1
|
Bai X, Zhu Q, Combs M, Wabitsch M, Mack CP, Taylor JM. GRAF1 deficiency leads to defective brown adipose tissue differentiation and thermogenic response. Sci Rep 2024; 14:28692. [PMID: 39562682 PMCID: PMC11577055 DOI: 10.1038/s41598-024-79301-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
Adipose tissue, which is crucial for the regulation of energy within the body, contains both white and brown adipocytes. White adipose tissue (WAT) primarily stores energy, while brown adipose tissue (BAT) plays a critical role in energy dissipation as heat, offering potential for therapies aimed at enhancing metabolic health. Regulation of the RhoA/ROCK pathway is crucial for appropriate specification, differentiation and maturation of both white and brown adipocytes. However, our knowledge of how this pathway is controlled within specific adipose depots remains unclear, and to date a RhoA regulator that selectively controls adipocyte browning has not been identified. Our study shows that GRAF1, a RhoGAP, is highly expressed in metabolically active tissues, and closely correlates with brown adipocyte differentiation in culture and in vivo. Mice with either global or adipocyte-specific GRAF1 deficiency exhibit impaired BAT maturation and compromised cold-induced thermogenesis. Moreover, defects in differentiation of human GRAF1-deficient brown preadipocytes can be rescued by treatment with a Rho kinase inhibitor. Collectively, these studies indicate that GRAF1 can selectively induce brown adipocyte differentiation and suggest that manipulating GRAF1 activity may hold promise for the future treatment of diseases related to metabolic dysfunction.
Collapse
Affiliation(s)
- Xue Bai
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Qiang Zhu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Matthew Combs
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Martin Wabitsch
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, 89075, Ulm, Germany
| | - Christopher P Mack
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
- McAllister Heart Institute, University of North Carolina, 160 North Medical Drive, 501 Brinkhous-Bullitt, CB# 7525, Chapel Hill, NC, 27599, USA
| | - Joan M Taylor
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.
- McAllister Heart Institute, University of North Carolina, 160 North Medical Drive, 501 Brinkhous-Bullitt, CB# 7525, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
2
|
Qiao E, Fulmore CA, Schaffer DV, Kumar S. Substrate stress relaxation regulates neural stem cell fate commitment. Proc Natl Acad Sci U S A 2024; 121:e2317711121. [PMID: 38968101 PMCID: PMC11252819 DOI: 10.1073/pnas.2317711121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 05/17/2024] [Indexed: 07/07/2024] Open
Abstract
Adult neural stem cells (NSCs) reside in the dentate gyrus of the hippocampus, and their capacity to generate neurons and glia plays a role in learning and memory. In addition, neurodegenerative diseases are known to be caused by a loss of neurons and glial cells, resulting in a need to better understand stem cell fate commitment processes. We previously showed that NSC fate commitment toward a neuronal or glial lineage is strongly influenced by extracellular matrix stiffness, a property of elastic materials. However, tissues in vivo are not purely elastic and have varying degrees of viscous character. Relatively little is known about how the viscoelastic properties of the substrate impact NSC fate commitment. Here, we introduce a polyacrylamide-based cell culture platform that incorporates mismatched DNA oligonucleotide-based cross-links as well as covalent cross-links. This platform allows for tunable viscous stress relaxation properties via variation in the number of mismatched base pairs. We find that NSCs exhibit increased astrocytic differentiation as the degree of stress relaxation is increased. Furthermore, culturing NSCs on increasingly stress-relaxing substrates impacts cytoskeletal dynamics by decreasing intracellular actin flow rates and stimulating cyclic activation of the mechanosensitive protein RhoA. Additionally, inhibition of motor-clutch model components such as myosin II and focal adhesion kinase partially or completely reverts cells to lineage distributions observed on elastic substrates. Collectively, our results introduce a unique system for controlling matrix stress relaxation properties and offer insight into how NSCs integrate viscoelastic cues to direct fate commitment.
Collapse
Affiliation(s)
- Eric Qiao
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA94720
| | - Camille A. Fulmore
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
| | - David V. Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- Department of Bioengineering, University of California, Berkeley, CA94720
| | - Sanjay Kumar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA94720
- Department of Bioengineering, University of California, Berkeley, CA94720
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA94143
| |
Collapse
|
3
|
Xie S, Liu Q, Fu C, Chen Y, Li M, Tian C, Li J, Han M, Li C. Molecular Regulation of Porcine Skeletal Muscle Development: Insights from Research on CDC23 Expression and Function. Int J Mol Sci 2024; 25:3664. [PMID: 38612477 PMCID: PMC11011816 DOI: 10.3390/ijms25073664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
Cell division cycle 23 (CDC23) is a component of the tetratricopeptide repeat (TPR) subunit in the anaphase-promoting complex or cyclosome (APC/C) complex, which participates in the regulation of mitosis in eukaryotes. However, the regulatory model and mechanism by which the CDC23 gene regulates muscle production in pigs are largely unknown. In this study, we investigated the expression of CDC23 in pigs, and the results indicated that CDC23 is widely expressed in various tissues and organs. In vitro cell experiments have demonstrated that CDC23 promotes the proliferation of myoblasts, as well as significantly positively regulating the differentiation of skeletal muscle satellite cells. In addition, Gene Set Enrichment Analysis (GSEA) revealed a significant downregulation of the cell cycle pathway during the differentiation process of skeletal muscle satellite cells. The protein-protein interaction (PPI) network showed a high degree of interaction between genes related to the cell cycle pathway and CDC23. Subsequently, in differentiated myocytes induced after overexpression of CDC23, the level of CDC23 exhibited a significant negative correlation with the expression of key factors in the cell cycle pathway, suggesting that CDC23 may be involved in the inhibition of the cell cycle signaling pathway in order to promote the differentiation process. In summary, we preliminarily determined the function of CDC23 with the aim of providing new insights into molecular regulation during porcine skeletal muscle development.
Collapse
Affiliation(s)
- Su Xie
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (S.X.); (Q.L.)
| | - Quan Liu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (S.X.); (Q.L.)
| | - Chong Fu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (S.X.); (Q.L.)
| | - Yansen Chen
- TERRA Teaching and Research Center, University of Liège, Gembloux Agro-Bio Tech (ULiège-GxABT), 5030 Gembloux, Belgium;
| | - Mengxun Li
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (S.X.); (Q.L.)
| | - Cheng Tian
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (S.X.); (Q.L.)
| | - Jiaxuan Li
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (S.X.); (Q.L.)
| | - Min Han
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (S.X.); (Q.L.)
| | - Changchun Li
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (S.X.); (Q.L.)
| |
Collapse
|
4
|
Noviello C, Kobon K, Randrianarison-Huetz V, Maire P, Pietri-Rouxel F, Falcone S, Sotiropoulos A. RhoA Is a Crucial Regulator of Myoblast Fusion. Cells 2023; 12:2673. [PMID: 38067102 PMCID: PMC10705320 DOI: 10.3390/cells12232673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 12/18/2023] Open
Abstract
Satellite cells (SCs) are adult muscle stem cells that are mobilized when muscle homeostasis is perturbed. Here we show that RhoA in SCs is indispensable to have correct muscle regeneration and hypertrophy. In particular, the absence of RhoA in SCs prevents a correct SC fusion both to other RhoA-deleted SCs (regeneration context) and to growing control myofibers (hypertrophy context). We demonstrated that RhoA is dispensable for SCs proliferation and differentiation; however, RhoA-deleted SCs have an inefficient movement even if their cytoskeleton assembly is not altered. Proliferative myoblast and differentiated myotubes without RhoA display a decreased expression of Chordin, suggesting a crosstalk between these genes for myoblast fusion regulation. These findings demonstrate the importance of RhoA in SC fusion regulation and its requirement to achieve an efficient skeletal muscle homeostasis restoration.
Collapse
Affiliation(s)
- Chiara Noviello
- Institut Cochin, Université de Paris, INSERM U1016, CNRS, F-75014 Paris, France (P.M.); (A.S.)
- Centre de Recherche en Myologie, Sorbonne Université, INSERM UMRS 974, Institut de Myologie, F-75013 Paris, France;
| | - Kassandra Kobon
- Institut Cochin, Université de Paris, INSERM U1016, CNRS, F-75014 Paris, France (P.M.); (A.S.)
| | | | - Pascal Maire
- Institut Cochin, Université de Paris, INSERM U1016, CNRS, F-75014 Paris, France (P.M.); (A.S.)
| | - France Pietri-Rouxel
- Centre de Recherche en Myologie, Sorbonne Université, INSERM UMRS 974, Institut de Myologie, F-75013 Paris, France;
| | - Sestina Falcone
- Centre de Recherche en Myologie, Sorbonne Université, INSERM UMRS 974, Institut de Myologie, F-75013 Paris, France;
| | - Athanassia Sotiropoulos
- Institut Cochin, Université de Paris, INSERM U1016, CNRS, F-75014 Paris, France (P.M.); (A.S.)
| |
Collapse
|
5
|
Nguyen MT, Dash R, Jeong K, Lee W. Role of Actin-Binding Proteins in Skeletal Myogenesis. Cells 2023; 12:2523. [PMID: 37947600 PMCID: PMC10650911 DOI: 10.3390/cells12212523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
Maintenance of skeletal muscle quantity and quality is essential to ensure various vital functions of the body. Muscle homeostasis is regulated by multiple cytoskeletal proteins and myogenic transcriptional programs responding to endogenous and exogenous signals influencing cell structure and function. Since actin is an essential component in cytoskeleton dynamics, actin-binding proteins (ABPs) have been recognized as crucial players in skeletal muscle health and diseases. Hence, dysregulation of ABPs leads to muscle atrophy characterized by loss of mass, strength, quality, and capacity for regeneration. This comprehensive review summarizes the recent studies that have unveiled the role of ABPs in actin cytoskeletal dynamics, with a particular focus on skeletal myogenesis and diseases. This provides insight into the molecular mechanisms that regulate skeletal myogenesis via ABPs as well as research avenues to identify potential therapeutic targets. Moreover, this review explores the implications of non-coding RNAs (ncRNAs) targeting ABPs in skeletal myogenesis and disorders based on recent achievements in ncRNA research. The studies presented here will enhance our understanding of the functional significance of ABPs and mechanotransduction-derived myogenic regulatory mechanisms. Furthermore, revealing how ncRNAs regulate ABPs will allow diverse therapeutic approaches for skeletal muscle disorders to be developed.
Collapse
Affiliation(s)
- Mai Thi Nguyen
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (K.J.)
| | - Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea;
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Republic of Korea
| | - Kyuho Jeong
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (K.J.)
| | - Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (K.J.)
- Channelopathy Research Center, Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Republic of Korea
| |
Collapse
|
6
|
Sidorenko E, Sokolova M, Pennanen AP, Kyheröinen S, Posern G, Foisner R, Vartiainen MK. Lamina-associated polypeptide 2α is required for intranuclear MRTF-A activity. Sci Rep 2022; 12:2306. [PMID: 35145145 PMCID: PMC8831594 DOI: 10.1038/s41598-022-06135-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/18/2022] [Indexed: 12/16/2022] Open
Abstract
Myocardin-related transcription factor A (MRTF-A), a coactivator of serum response factor (SRF), regulates the expression of many cytoskeletal genes in response to cytoplasmic and nuclear actin dynamics. Here we describe a novel mechanism to regulate MRTF-A activity within the nucleus by showing that lamina-associated polypeptide 2α (Lap2α), the nucleoplasmic isoform of Lap2, is a direct binding partner of MRTF-A, and required for the efficient expression of MRTF-A/SRF target genes. Mechanistically, Lap2α is not required for MRTF-A nuclear localization, unlike most other MRTF-A regulators, but is required for efficient recruitment of MRTF-A to its target genes. This regulatory step takes place prior to MRTF-A chromatin binding, because Lap2α neither interacts with, nor specifically influences active histone marks on MRTF-A/SRF target genes. Phenotypically, Lap2α is required for serum-induced cell migration, and deregulated MRTF-A activity may also contribute to muscle and proliferation phenotypes associated with loss of Lap2α. Our studies therefore add another regulatory layer to the control of MRTF-A-SRF-mediated gene expression, and broaden the role of Lap2α in transcriptional regulation.
Collapse
Affiliation(s)
| | - Maria Sokolova
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Antti P Pennanen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Salla Kyheröinen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Guido Posern
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Roland Foisner
- Max Perutz Labs, Center for Medical Biochemistry, Medical University of Vienna, Vienna Biocenter Campus (VBC), Vienna, Austria
| | | |
Collapse
|
7
|
Szabo K, Varga D, Vegh AG, Liu N, Xiao X, Xu L, Dux L, Erdelyi M, Rovo L, Keller-Pinter A. Syndecan-4 affects myogenesis via Rac1-mediated actin remodeling and exhibits copy-number amplification and increased expression in human rhabdomyosarcoma tumors. Cell Mol Life Sci 2022; 79:122. [PMID: 35128576 PMCID: PMC8818642 DOI: 10.1007/s00018-021-04121-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/14/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Skeletal muscle demonstrates a high degree of regenerative capacity repeating the embryonic myogenic program under strict control. Rhabdomyosarcoma is the most common sarcoma in childhood and is characterized by impaired muscle differentiation. In this study, we observed that silencing the expression of syndecan-4, the ubiquitously expressed transmembrane heparan sulfate proteoglycan, significantly enhanced myoblast differentiation, and fusion. During muscle differentiation, the gradually decreasing expression of syndecan-4 allows the activation of Rac1, thereby mediating myoblast fusion. Single-molecule localized superresolution direct stochastic optical reconstruction microscopy (dSTORM) imaging revealed nanoscale changes in actin cytoskeletal architecture, and atomic force microscopy showed reduced elasticity of syndecan-4-knockdown cells during fusion. Syndecan-4 copy-number amplification was observed in 28% of human fusion-negative rhabdomyosarcoma tumors and was accompanied by increased syndecan-4 expression based on RNA sequencing data. Our study suggests that syndecan-4 can serve as a tumor driver gene in promoting rabdomyosarcoma tumor development. Our results contribute to the understanding of the role of syndecan-4 in skeletal muscle development, regeneration, and tumorigenesis.
Collapse
|
8
|
Lv Z, Ding Y, Cao W, Wang S, Gao K. Role of RHO family interacting cell polarization regulators (RIPORs) in health and disease: Recent advances and prospects. Int J Biol Sci 2022; 18:800-808. [PMID: 35002526 PMCID: PMC8741841 DOI: 10.7150/ijbs.65457] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/27/2021] [Indexed: 12/11/2022] Open
Abstract
The RHO GTPase family has been suggested to play critical roles in cell growth, migration, and polarization. Regulators and effectors of RHO GTPases have been extensively explored in recent years. However, little attention has been given to RHO family interacting cell polarization regulators (RIPORs), a recently discovered protein family of RHO regulators. RIPOR proteins, namely, RIPOR1-3, bind directly to RHO proteins (A, B and C) via a RHO-binding motif and exert suppressive effects on RHO activity, thereby negatively influencing RHO-regulated cellular functions. In addition, RIPORs are phosphorylated by upstream protein kinases under chemokine stimulation, and this phosphorylation affects not only their subcellular localization but also their interaction with RHO proteins, altering the activation of RHO downstream targets and ultimately impacting cell polarity and migration. In this review, we provide an overview of recent studies on the function of RIPOR proteins in regulating RHO-dependent directional movement in immune responses and other pathophysiological functions.
Collapse
Affiliation(s)
- Zeheng Lv
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yan Ding
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Wenxin Cao
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Shuyun Wang
- Department of Breast Surgery, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Kun Gao
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| |
Collapse
|
9
|
Hammers DW, Hart CC, Matheny MK, Heimsath EG, Lee YI, Hammer JA, Cheney RE, Sweeney HL. Filopodia powered by class x myosin promote fusion of mammalian myoblasts. eLife 2021; 10:e72419. [PMID: 34519272 PMCID: PMC8500716 DOI: 10.7554/elife.72419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/13/2021] [Indexed: 12/30/2022] Open
Abstract
Skeletal muscle fibers are multinucleated cellular giants formed by the fusion of mononuclear myoblasts. Several molecules involved in myoblast fusion have been discovered, and finger-like projections coincident with myoblast fusion have also been implicated in the fusion process. The role of these cellular projections in muscle cell fusion was investigated herein. We demonstrate that these projections are filopodia generated by class X myosin (Myo10), an unconventional myosin motor protein specialized for filopodia. We further show that Myo10 is highly expressed by differentiating myoblasts, and Myo10 ablation inhibits both filopodia formation and myoblast fusion in vitro. In vivo, Myo10 labels regenerating muscle fibers associated with Duchenne muscular dystrophy and acute muscle injury. In mice, conditional loss of Myo10 from muscle-resident stem cells, known as satellite cells, severely impairs postnatal muscle regeneration. Furthermore, the muscle fusion proteins Myomaker and Myomixer are detected in myoblast filopodia. These data demonstrate that Myo10-driven filopodia facilitate multinucleated mammalian muscle formation.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Cell Fusion
- Cell Line
- Cell Proliferation
- Disease Models, Animal
- Humans
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Mice, Knockout
- Muscle Development
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Myoblasts, Skeletal/metabolism
- Myoblasts, Skeletal/pathology
- Myosins/genetics
- Myosins/metabolism
- Pseudopodia/genetics
- Pseudopodia/metabolism
- Regeneration
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/pathology
- Time Factors
- Mice
Collapse
Affiliation(s)
- David W Hammers
- Department of Pharmacology & Therapeutics, University of Florida College of MedicineGainesvilleUnited States
- University of Florida Myology InstituteGainesvilleUnited States
| | - Cora C Hart
- Department of Pharmacology & Therapeutics, University of Florida College of MedicineGainesvilleUnited States
- University of Florida Myology InstituteGainesvilleUnited States
| | - Michael K Matheny
- Department of Pharmacology & Therapeutics, University of Florida College of MedicineGainesvilleUnited States
- University of Florida Myology InstituteGainesvilleUnited States
| | - Ernest G Heimsath
- Department of Cell Biology & Physiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of MedicineChapel HillUnited States
| | - Young il Lee
- Department of Pharmacology & Therapeutics, University of Florida College of MedicineGainesvilleUnited States
- University of Florida Myology InstituteGainesvilleUnited States
| | - John A Hammer
- Cell Biology and Physiology Center, National Heart, Lung and Blood InstituteBethesdaUnited States
| | - Richard E Cheney
- Department of Cell Biology & Physiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of MedicineChapel HillUnited States
| | - H Lee Sweeney
- Department of Pharmacology & Therapeutics, University of Florida College of MedicineGainesvilleUnited States
- University of Florida Myology InstituteGainesvilleUnited States
| |
Collapse
|
10
|
Kurosaka M, Ogura Y, Sato S, Kohda K, Funabashi T. Transcription factor signal transducer and activator of transcription 6 (STAT6) is an inhibitory factor for adult myogenesis. Skelet Muscle 2021; 11:14. [PMID: 34051858 PMCID: PMC8164270 DOI: 10.1186/s13395-021-00271-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 05/18/2021] [Indexed: 01/25/2023] Open
Abstract
Background The signal transducer and activator of transcription 6 (STAT6) transcription factor plays a vitally important role in immune cells, where it is activated mainly by interleukin-4 (IL-4). Because IL-4 is an essential cytokine for myotube formation, STAT6 might also be involved in myogenesis as part of IL-4 signaling. This study was conducted to elucidate the role of STAT6 in adult myogenesis in vitro and in vivo. Methods Myoblasts were isolated from male mice and were differentiated on a culture dish to evaluate the change in STAT6 during myotube formation. Then, the effects of STAT6 overexpression and inhibition on proliferation, differentiation, and fusion in those cells were studied. Additionally, to elucidate the myogenic role of STAT6 in vivo, muscle regeneration after injury was evaluated in STAT6 knockout mice. Results IL-4 can increase STAT6 phosphorylation, but STAT6 phosphorylation decreased during myotube formation in culture. STAT6 overexpression decreased, but STAT6 knockdown increased the differentiation index and the fusion index. Results indicate that STAT6 inhibited myogenin protein expression. Results of in vivo experiments show that STAT6 knockout mice exhibited better regeneration than wild-type mice 5 days after cardiotoxin-induced injury. It is particularly interesting that results obtained using cells from STAT6 knockout mice suggest that this STAT6 inhibitory action for myogenesis was not mediated by IL-4 but might instead be associated with p38 mitogen-activated protein kinase phosphorylation. However, STAT6 was not involved in the proliferation of myogenic cells in vitro and in vivo. Conclusion Results suggest that STAT6 functions as an inhibitor of adult myogenesis. Moreover, results suggest that the IL-4-STAT6 signaling axis is unlikely to be responsible for myotube formation. Supplementary Information The online version contains supplementary material available at 10.1186/s13395-021-00271-8.
Collapse
Affiliation(s)
- Mitsutoshi Kurosaka
- Department of Physiology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan
| | - Yuji Ogura
- Department of Physiology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan.
| | - Shuichi Sato
- School of Kinesiology, The University of Louisiana at Lafayette, Lafayette, LA, USA.,New Iberia Research Center, The University of Louisiana at Lafayette, New Iberia, LA, USA
| | - Kazuhisa Kohda
- Department of Physiology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan
| | - Toshiya Funabashi
- Department of Physiology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan
| |
Collapse
|
11
|
Chen M, Zhang L, Guo Y, Liu X, Song Y, Li X, Ding X, Guo H. A novel lncRNA promotes myogenesis of bovine skeletal muscle satellite cells via PFN1-RhoA/Rac1. J Cell Mol Med 2021; 25:5988-6005. [PMID: 33942976 PMCID: PMC8256363 DOI: 10.1111/jcmm.16427] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
Myogenesis, the process of skeletal muscle formation, is a highly coordinated multistep biological process. Accumulating evidence suggests that long non-coding RNAs (lncRNAs) are emerging as a gatekeeper in myogenesis. Up to now, most studies on muscle development-related lncRNAs are mainly focussed on humans and mice. In this study, a novel muscle highly expressed lncRNA, named lnc23, localized in nucleus, was found differentially expressed in different stages of embryonic development and myogenic differentiation. The knockdown and over-expression experiments showed that lnc23 positively regulated the myogenic differentiation of bovine skeletal muscle satellite cells. Then, TMT 10-plex labelling quantitative proteomics was performed to screen the potentially regulatory proteins of lnc23. Results indicated that lnc23 was involved in the key processes of myogenic differentiation such as cell fusion, further demonstrated that down-regulation of lnc23 may inhibit myogenic differentiation by reducing signal transduction and cell fusion among cells. Furthermore, RNA pulldown/LC-MS and RIP experiment illustrated that PFN1 was a binding protein of lnc23. Further, we also found that lnc23 positively regulated the protein expression of RhoA and Rac1, and PFN1 may negatively regulate myogenic differentiation and the expression of its interacting proteins RhoA and Rac1. Hence, we support that lnc23 may reduce the inhibiting effect of PFN1 on RhoA and Rac1 by binding to PFN1, thereby promoting myogenic differentiation. In short, the novel identified lnc23 promotes myogenesis of bovine skeletal muscle satellite cells via PFN1-RhoA/Rac1.
Collapse
Affiliation(s)
- Mingming Chen
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy HusbandryCollege of Animal Science and Veterinary MedicineTianjin Agricultural UniversityTianjinChina
| | - Linlin Zhang
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy HusbandryCollege of Animal Science and Veterinary MedicineTianjin Agricultural UniversityTianjinChina
| | - Yiwen Guo
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy HusbandryCollege of Animal Science and Veterinary MedicineTianjin Agricultural UniversityTianjinChina
| | - Xinfeng Liu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy HusbandryCollege of Animal Science and Veterinary MedicineTianjin Agricultural UniversityTianjinChina
| | - Yingshen Song
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy HusbandryCollege of Animal Science and Veterinary MedicineTianjin Agricultural UniversityTianjinChina
| | - Xin Li
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy HusbandryCollege of Animal Science and Veterinary MedicineTianjin Agricultural UniversityTianjinChina
| | - Xiangbin Ding
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy HusbandryCollege of Animal Science and Veterinary MedicineTianjin Agricultural UniversityTianjinChina
| | - Hong Guo
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy HusbandryCollege of Animal Science and Veterinary MedicineTianjin Agricultural UniversityTianjinChina
| |
Collapse
|
12
|
The cooperation of cis-elements during M-cadherin promoter activation. Biochem J 2021; 478:911-926. [PMID: 33527978 DOI: 10.1042/bcj20200535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 01/21/2021] [Accepted: 02/02/2021] [Indexed: 01/26/2023]
Abstract
M-cadherin is a skeletal muscle-specific transmembrane protein mediating the cell-cell adhesion of myoblasts during myogenesis. It is expressed in the proliferating satellite cells and highly induced by myogenic regulatory factors (MRFs) during terminal myogenic differentiation. Several conserved cis-elements, including 5 E-boxes, 2 GC boxes, and 1 conserved downstream element (CDE) were identified in the M-cadherin proximal promoter. We found that E-box-3 and -4 close to the transcription initiation site (TIS) mediated most of its transactivation by MyoD, the strongest myogenic MRF. Including of any one of the other E-boxes restored the full activation by MyoD, suggesting an essential collaboration between E-boxes. Stronger activation of M-cadherin promoter than that of muscle creatine kinase (MCK) by MyoD was observed regardless of culture conditions and the presence of E47. Furthermore, MyoD/E47 heterodimer and MyoD ∼ E47 fusion protein achieved similar levels of activation in differentiation medium (DM), suggesting high affinity of MyoD/E47 to E-boxes 3/4 under DM. We also found that GC boxes and CDE positively affected MyoD mediated activation. The CDE element was predicted to be the target of the chromatin-modifying factor Meis1/Pbx1 heterodimer. Knockdown of Pbx1 significantly reduced the expression level of M-cadherin, but increased that of N-cadherin. Using ChIP assay, we further found significant reduction in MyoD recruitment to M-cadherin promoter when CDE was deleted. Taken together, these observations suggest that the chromatin-modifying function of Pbx1/Meis1 is critical to M-cadherin promoter activation before MyoD is recruited to E-boxes to trigger transcription.
Collapse
|
13
|
You JS, Singh N, Reyes-Ordonez A, Khanna N, Bao Z, Zhao H, Chen J. ARHGEF3 Regulates Skeletal Muscle Regeneration and Strength through Autophagy. Cell Rep 2021; 34:108594. [PMID: 33406419 DOI: 10.1016/j.celrep.2020.108594] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 09/15/2020] [Accepted: 12/10/2020] [Indexed: 12/18/2022] Open
Abstract
Skeletal muscle regeneration after injury is essential for maintaining muscle function throughout aging. ARHGEF3, a RhoA/B-specific GEF, negatively regulates myoblast differentiation through Akt signaling independently of its GEF activity in vitro. Here, we report ARHGEF3's role in skeletal muscle regeneration revealed by ARHGEF3-KO mice. These mice exhibit indiscernible phenotype under basal conditions. Upon acute injury, however, ARHGEF3 deficiency enhances the mass/fiber size and function of regenerating muscles in both young and regeneration-defective middle-aged mice. Surprisingly, these effects occur independently of Akt but via the GEF activity of ARHGEF3. Consistently, overexpression of ARHGEF3 inhibits muscle regeneration in a Rho-associated kinase-dependent manner. We further show that ARHGEF3 KO promotes muscle regeneration through activation of autophagy, a process that is also critical for maintaining muscle strength. Accordingly, ARHGEF3 depletion in old mice prevents muscle weakness by restoring autophagy. Taken together, our findings identify a link between ARHGEF3 and autophagy-related muscle pathophysiology.
Collapse
Key Words
- Skeletal muscle, Regeneration, Aging, Strength, Muscle quality, ARHGEF3, XPLN, Akt, RhoA, ROCK, Autophagy
Collapse
Affiliation(s)
- Jae-Sung You
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Nilmani Singh
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Adriana Reyes-Ordonez
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Nidhi Khanna
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Modalis Therapeutics, Cambridge, MA 02138, USA
| | - Zehua Bao
- Department of Chemical and Biomolecular Engineering and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, MA 02215, USA
| | - Huimin Zhao
- Department of Chemical and Biomolecular Engineering and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jie Chen
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
14
|
Mu X, Lin CY, Hambright WS, Tang Y, Ravuri S, Lu A, Matre P, Chen W, Gao X, Cui Y, Zhong L, Wang B, Huard J. Aberrant RhoA activation in macrophages increases senescence-associated secretory phenotypes and ectopic calcification in muscular dystrophic mice. Aging (Albany NY) 2020; 12:24853-24871. [PMID: 33361519 PMCID: PMC7803538 DOI: 10.18632/aging.202413] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 11/21/2020] [Indexed: 12/17/2022]
Abstract
Duchenne Muscular Dystrophy (DMD) patients often suffer from both muscle wasting and osteoporosis. Our previous studies have revealed reduced regeneration potential in skeletal muscle and bone, concomitant with ectopic calcification of soft tissues in double knockout (dKO, dystrophin-/-; utrophin-/-) mice, a severe murine model for DMD. We found significant involvement of RhoA/ROCK (Rho-Associated Protein Kinase) signaling in mediating ectopic calcification of muscles in dKO mice. However, the cellular identity of these RhoA+ cells, and the role that RhoA plays in the chronic inflammation-associated pathologies has not been elucidated. Here, we report that CD68+ macrophages are highly prevalent at the sites of ectopic calcification of dKO mice, and that these macrophages highly express RhoA. Macrophages from dKO mice feature a shift towards a more pro-inflammatory M1 polarization and an increased expression of various senescence-associated secretory phenotype (SASP) factors that was reduced with the RhoA/ROCK inhibitor Y-27632. Further, systemic inhibition of RhoA activity in dKO mice led to reduced number of RhoA+/CD68+ cells, as well as a reduction in fibrosis and ectopic calcification. Together, these data revealed that RhoA signaling may be a key regulator of imbalanced mineralization in the dystrophic musculoskeletal system and consequently a therapeutic target for the treatment of DMD or other related muscle dystrophies.
Collapse
Affiliation(s)
- Xiaodong Mu
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.,Steadman Philippon Research Institute, Center for Regenerative Sports Medicine, Vail, CO 81657, USA
| | - Chi-Yi Lin
- Department of Orthopedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - William S Hambright
- Steadman Philippon Research Institute, Center for Regenerative Sports Medicine, Vail, CO 81657, USA
| | - Ying Tang
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Sudheer Ravuri
- Steadman Philippon Research Institute, Center for Regenerative Sports Medicine, Vail, CO 81657, USA
| | - Aiping Lu
- Steadman Philippon Research Institute, Center for Regenerative Sports Medicine, Vail, CO 81657, USA
| | - Polina Matre
- Department of Orthopedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Wanqun Chen
- Department of Orthopedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,Department of Biochemistry and Molecular Biology, Jinan University, Guangzhou, China
| | - Xueqin Gao
- Steadman Philippon Research Institute, Center for Regenerative Sports Medicine, Vail, CO 81657, USA
| | - Yan Cui
- Department of Orthopedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ling Zhong
- Department of Orthopedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Bing Wang
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Johnny Huard
- Steadman Philippon Research Institute, Center for Regenerative Sports Medicine, Vail, CO 81657, USA
| |
Collapse
|
15
|
Holstein I, Singh AK, Pohl F, Misiak D, Braun J, Leitner L, Hüttelmaier S, Posern G. Post-transcriptional regulation of MRTF-A by miRNAs during myogenic differentiation of myoblasts. Nucleic Acids Res 2020; 48:8927-8942. [PMID: 32692361 PMCID: PMC7498330 DOI: 10.1093/nar/gkaa596] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 07/01/2020] [Accepted: 07/09/2020] [Indexed: 12/02/2022] Open
Abstract
The differentiation and regeneration of skeletal muscle from myoblasts to myotubes involves myogenic transcription factors, such as myocardin-related transcription factor A (MRTF-A) and serum response factor (SRF). In addition, post-transcriptional regulation by miRNAs is required during myogenesis. Here, we provide evidence for novel mechanisms regulating MRTF-A during myogenic differentiation. Endogenous MRTF-A protein abundance and activity decreased during C2C12 differentiation, which was attributable to miRNA-directed inhibition. Conversely, overexpression of MRTF-A impaired differentiation and myosin expression. Applying miRNA trapping by RNA affinity purification (miTRAP), we identified miRNAs which directly regulate MRTF-A via its 3′UTR, including miR-1a-3p, miR-206-3p, miR-24-3p and miR-486-5p. These miRNAs were upregulated during differentiation and specifically recruited to the 3′UTR of MRTF-A. Concomitantly, Ago2 recruitment to the MRTF-A 3′UTR was considerably increased, whereas Dicer1 depletion or 3′UTR deletion elevated MRTF-A and inhibited differentiation. MRTF-A protein expression was inhibited by ectopic miRNA expression in murine C2C12 and primary human myoblasts. 3′UTR reporter activity diminished upon differentiation or miRNA expression, whereas deletion of the predicted binding sites reversed these effects. Furthermore, TGF-β abolished MRTF-A reduction and decreased miR-486-5p expression. Our findings implicate miR-24-3p and miR-486-5p in the repression of MRTF-A and suggest a complex network of transcriptional and post-transcriptional mechanisms regulating myogenesis.
Collapse
Affiliation(s)
- Ingo Holstein
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, Hollystrasse 1, 06114 Halle (Saale), Germany
| | - Anurag Kumar Singh
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, Hollystrasse 1, 06114 Halle (Saale), Germany
| | - Falk Pohl
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, Hollystrasse 1, 06114 Halle (Saale), Germany
| | - Danny Misiak
- Institute of Molecular Medicine, Medical Faculty, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Center, Kurt-Mothes-Straße 3a, 06120 Halle (Saale), Germany
| | - Juliane Braun
- Institute of Molecular Medicine, Medical Faculty, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Center, Kurt-Mothes-Straße 3a, 06120 Halle (Saale), Germany
| | - Laura Leitner
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, Hollystrasse 1, 06114 Halle (Saale), Germany
| | - Stefan Hüttelmaier
- Institute of Molecular Medicine, Medical Faculty, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Center, Kurt-Mothes-Straße 3a, 06120 Halle (Saale), Germany
| | - Guido Posern
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, Hollystrasse 1, 06114 Halle (Saale), Germany
| |
Collapse
|
16
|
Liu Q, Yu F, Yi L, Gao Y, Gui R, Yi M, Sun J. Stochastic Dynamics of Gene Switching and Energy Dissipation for Gene Expression. Front Genet 2020; 11:676. [PMID: 32714375 PMCID: PMC7343763 DOI: 10.3389/fgene.2020.00676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/02/2020] [Indexed: 11/19/2022] Open
Abstract
Stochastic dynamics of gene switching and energy dissipation for gene expression are largely unknown, mainly due to the complexity of non-equilibrium mechanisms. Here, based on an important double-deck loop model, the stochastic mechanisms of gene switching and energy dissipation are studied. First, the probability distributions of steady states are calculated theoretically. It is found that the signal can strengthen the choice of gene switching between the "off" and "on" states. Our analysis of energy consumption illustrates that, compared with the synthesis and degradation of proteins, the process of gene switching costs little energy. Our theoretical analysis reveals some interesting insights into the determination of cell state and energy dissipation for gene expression.
Collapse
Affiliation(s)
- Quan Liu
- Department of Physics, College of Science, Huazhong Agricultural University, Wuhan, China
| | - FengZhen Yu
- Department of Chemistry, College of Science, Huazhong Agricultural University, Wuhan, China
| | - Liang Yi
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yijun Gao
- Department of Physics, College of Science, Huazhong Agricultural University, Wuhan, China
| | - Rong Gui
- Department of Physics, College of Science, Huazhong Agricultural University, Wuhan, China
| | - Ming Yi
- School of Mathematics and Physics, China University of Geosciences, Wuhan, China
| | - Jianqiang Sun
- School of Automation and Electrical Engineering, Linyi University, Linyi, China
| |
Collapse
|
17
|
Chen B, You W, Wang Y, Shan T. The regulatory role of Myomaker and Myomixer-Myomerger-Minion in muscle development and regeneration. Cell Mol Life Sci 2020; 77:1551-1569. [PMID: 31642939 PMCID: PMC11105057 DOI: 10.1007/s00018-019-03341-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022]
Abstract
Skeletal muscle plays essential roles in motor function, energy, and glucose metabolism. Skeletal muscle formation occurs through a process called myogenesis, in which a crucial step is the fusion of mononucleated myoblasts to form multinucleated myofibers. The myoblast/myocyte fusion is triggered and coordinated in a muscle-specific way that is essential for muscle development and post-natal muscle regeneration. Many molecules and proteins have been found and demonstrated to have the capacity to regulate the fusion of myoblast/myocytes. Interestingly, two newly discovered muscle-specific membrane proteins, Myomaker and Myomixer (also called Myomerger and Minion), have been identified as fusogenic regulators in vertebrates. Both Myomaker and Myomixer-Myomerger-Minion have the capacity to directly control the myogenic fusion process. Here, we review and discuss the latest studies related to these two proteins, including the discovery, structure, expression pattern, functions, and regulation of Myomaker and Myomixer-Myomerger-Minion. We also emphasize and discuss the interaction between Myomaker and Myomixer-Myomerger-Minion, as well as their cooperative regulatory roles in cell-cell fusion. Moreover, we highlight the areas for exploration of Myomaker and Myomixer-Myomerger-Minion in future studies and consider their potential application to control cell fusion for cell-therapy purposes.
Collapse
Affiliation(s)
- Bide Chen
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Wenjing You
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Yizhen Wang
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, China.
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China.
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China.
| |
Collapse
|
18
|
Carrero-Rojas G, Benítez-Temiño B, Pastor AM, Davis López de Carrizosa MA. Muscle Progenitors Derived from Extraocular Muscles Express Higher Levels of Neurotrophins and their Receptors than other Cranial and Limb Muscles. Cells 2020; 9:cells9030747. [PMID: 32197508 PMCID: PMC7140653 DOI: 10.3390/cells9030747] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/02/2020] [Accepted: 03/17/2020] [Indexed: 01/19/2023] Open
Abstract
Extraocular muscles (EOMs) show resistance to muscle dystrophies and sarcopenia. It has been recently demonstrated that they are endowed with different types of myogenic cells, all of which present an outstanding regenerative potential. Neurotrophins are important modulators of myogenic regeneration and act promoting myoblast proliferation, enhancing myogenic fusion rates and protecting myotubes from inflammatory stimuli. Here, we adapted the pre-plate cell isolation technique to obtain myogenic progenitors from the rat EOMs, and quantified their in vitro expression of neurotrophins and their receptors by RT–qPCR and immunohistochemistry, respectively. The results were compared with the expression on progenitors isolated from buccinator, tongue and limb muscles. Our quantitative analysis of brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF) and neurotrophin-3 (NT-3) transcripts showed, for the first time, that EOMs-derived cells express more of these factors and that they expressed TrkA, but not TrkB and TrkC receptors. On the contrary, the immunofluorescence analysis demonstrated high expression of p75NTR on all myogenic progenitors, with the EOMs-derived cells showing higher expression. Taken together, these results suggest that the intrinsic trophic differences between EOMs-derived myogenic progenitors and their counterparts from other muscles could explain why those cells show higher proliferative and fusion rates, as well as better regenerative properties.
Collapse
|
19
|
Mierzejewski B, Archacka K, Grabowska I, Florkowska A, Ciemerych MA, Brzoska E. Human and mouse skeletal muscle stem and progenitor cells in health and disease. Semin Cell Dev Biol 2020; 104:93-104. [PMID: 32005567 DOI: 10.1016/j.semcdb.2020.01.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/15/2020] [Accepted: 01/15/2020] [Indexed: 12/25/2022]
Abstract
The proper functioning of tissues and organs depends on their ability to self-renew and repair. Some of the tissues, like epithelia, renew almost constantly while in the others this process is induced by injury or diseases. The stem or progenitor cells responsible for tissue homeostasis have been identified in many organs. Some of them, such as hematopoietic or intestinal epithelium stem cells, are multipotent and can differentiate into various cell types. Others are unipotent. The skeletal muscle tissue does not self-renew spontaneously, however, it presents unique ability to regenerate in response to the injury or disease. Its repair almost exclusively relies on unipotent satellite cells. However, multiple lines of evidence document that some progenitor cells present in the muscle can be supportive for skeletal muscle regeneration. Here, we summarize the current knowledge on the complicated landscape of stem and progenitor cells that exist in skeletal muscle and support its regeneration. We compare the cells from two model organisms, i.e., mouse and human, documenting their similarities and differences and indicating methods to test their ability to undergo myogenic differentiation.
Collapse
Affiliation(s)
- Bartosz Mierzejewski
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1St, 02-096 Warsaw, Poland
| | - Karolina Archacka
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1St, 02-096 Warsaw, Poland
| | - Iwona Grabowska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1St, 02-096 Warsaw, Poland
| | - Anita Florkowska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1St, 02-096 Warsaw, Poland
| | - Maria Anna Ciemerych
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1St, 02-096 Warsaw, Poland
| | - Edyta Brzoska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1St, 02-096 Warsaw, Poland.
| |
Collapse
|
20
|
Figeac N, Pruller J, Hofer I, Fortier M, Ortuste Quiroga HP, Banerji CRS, Zammit PS. DEPDC1B is a key regulator of myoblast proliferation in mouse and man. Cell Prolif 2020; 53:e12717. [PMID: 31825138 PMCID: PMC6985657 DOI: 10.1111/cpr.12717] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/19/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES DISHEVELLED, EGL-10, PLECKSTRIN (DEP) domain-containing 1B (DEPDC1B) promotes dismantling of focal adhesions and coordinates detachment events during cell cycle progression. DEPDC1B is overexpressed in several cancers with expression inversely correlated with patient survival. Here, we analysed the role of DEPDC1B in the regulation of murine and human skeletal myogenesis. MATERIALS AND METHODS Expression dynamics of DEPDC1B were examined in murine and human myoblasts and rhabdomyosarcoma cells in vitro by RT-qPCR and/or immunolabelling. DEPDC1B function was mainly tested via siRNA-mediated gene knockdown. RESULTS DEPDC1B was expressed in proliferating murine and human myoblasts, with expression then decreasing markedly during myogenic differentiation. SiRNA-mediated knockdown of DEPDC1B reduced myoblast proliferation and induced entry into myogenic differentiation, with deregulation of key cell cycle regulators (cyclins, CDK, CDKi). DEPDC1B and β-catenin co-knockdown was unable to rescue proliferation in myoblasts, suggesting that DEPDC1B functions independently of canonical WNT signalling during myogenesis. DEPDC1B can also suppress RHOA activity in some cell types, but DEPDC1B and RHOA co-knockdown actually had an additive effect by both further reducing proliferation and enhancing myogenic differentiation. DEPDC1B was expressed in human Rh30 rhabdomyosarcoma cells, where DEPDC1B or RHOA knockdown promoted myogenic differentiation, but without influencing proliferation. CONCLUSION DEPDC1B plays a central role in myoblasts by driving proliferation and preventing precocious myogenic differentiation during skeletal myogenesis in both mouse and human.
Collapse
Affiliation(s)
- Nicolas Figeac
- King's College LondonRandall Centre for Cell and Molecular BiophysicsLondonUK
| | - Johanna Pruller
- King's College LondonRandall Centre for Cell and Molecular BiophysicsLondonUK
| | - Isabella Hofer
- King's College LondonRandall Centre for Cell and Molecular BiophysicsLondonUK
| | - Mathieu Fortier
- King's College LondonRandall Centre for Cell and Molecular BiophysicsLondonUK
| | | | | | - Peter S. Zammit
- King's College LondonRandall Centre for Cell and Molecular BiophysicsLondonUK
| |
Collapse
|
21
|
Actomyosin contractility scales with myoblast elongation and enhances differentiation through YAP nuclear export. Sci Rep 2019; 9:15565. [PMID: 31664178 PMCID: PMC6820726 DOI: 10.1038/s41598-019-52129-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 10/10/2019] [Indexed: 01/14/2023] Open
Abstract
Skeletal muscle fibers are formed by the fusion of mononucleated myoblasts into long linear myotubes, which differentiate and reorganize into multinucleated myofibers that assemble in bundles to form skeletal muscles. This fundamental process requires the elongation of myoblasts into a bipolar shape, although a complete understanding of the mechanisms governing skeletal muscle fusion is lacking. To address this question, we consider cell aspect ratio, actomyosin contractility and the Hippo pathway member YAP as potential regulators of the fusion of myoblasts into myotubes. Using fibronectin micropatterns of different geometries and traction force microscopy, we investigated how myoblast elongation affects actomyosin contractility. Our findings indicate that cell elongation enhances actomyosin contractility in myoblasts, which regulate their actin network to their spreading area. Interestingly, we found that the contractility of cell pairs increased after their fusion and raise on elongated morphologies. Furthermore, our findings indicate that myoblast elongation modulates nuclear orientation and triggers cytoplasmic localization of YAP, increasing evidence that YAP is a key regulator of mechanotransduction in myoblasts. Taken together, our findings support a mechanical model where actomyosin contractility scales with myoblast elongation and enhances the differentiation of myoblasts into myotubes through YAP nuclear export.
Collapse
|
22
|
Dimartino D, Colantoni A, Ballarino M, Martone J, Mariani D, Danner J, Bruckmann A, Meister G, Morlando M, Bozzoni I. The Long Non-coding RNA lnc-31 Interacts with Rock1 mRNA and Mediates Its YB-1-Dependent Translation. Cell Rep 2019; 23:733-740. [PMID: 29669280 PMCID: PMC5917449 DOI: 10.1016/j.celrep.2018.03.101] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/20/2017] [Accepted: 03/21/2018] [Indexed: 01/08/2023] Open
Abstract
Cytoplasmic long non-coding RNAs have been shown to act at many different levels to control post-transcriptional gene expression, although their role in translational control is poorly understood. Here, we show that lnc-31, a non-coding RNA required for myoblast proliferation, promotes ROCK1 protein synthesis by stabilizing its translational activator, YB-1. We find that lnc-31 binds to the Rock1 mRNA as well as to the YB-1 protein and that translational activation requires physical interaction between the two RNA species. These results suggest a localized effect of YB-1 stabilization on the Rock1 mRNA. ROCK1 upregulation by lnc-31, in proliferative conditions, correlates well with the differentiation-repressing activity of ROCK1. We also show that, upon induction of differentiation, the downregulation of lnc-31, in conjunction with miR-152 targeting of Rock1, establishes a regulatory loop that reinforces ROCK1 repression and promotes myogenesis. lnc-31 sustains myoblast proliferation, counteracting differentiation lnc-31 binds to Rock1 mRNA and YB-1 protein Rock-1 translation is favored through its interaction with lnc-31 and YB-1 protein lnc-31 counteracts YB-1 protein degradation, thus promoting Rock1 translation
Collapse
Affiliation(s)
- Dacia Dimartino
- Department of Biology and Biotechnology, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Alessio Colantoni
- Department of Biology and Biotechnology, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Monica Ballarino
- Department of Biology and Biotechnology, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Julie Martone
- Department of Biology and Biotechnology, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Davide Mariani
- Department of Biology and Biotechnology, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Johannes Danner
- Biochemistry Center Regensburg, Laboratory for RNA Biology, University of Regensburg, 93053 Regensburg, Germany
| | - Astrid Bruckmann
- Biochemistry Center Regensburg, Laboratory for RNA Biology, University of Regensburg, 93053 Regensburg, Germany
| | - Gunter Meister
- Biochemistry Center Regensburg, Laboratory for RNA Biology, University of Regensburg, 93053 Regensburg, Germany
| | - Mariangela Morlando
- Department of Biology and Biotechnology, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| | - Irene Bozzoni
- Department of Biology and Biotechnology, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy; Institute Pasteur Fondazione Cenci-Bolognetti, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| |
Collapse
|
23
|
Saleh A, Subramaniam G, Raychaudhuri S, Dhawan J. Cytoplasmic sequestration of the RhoA effector mDiaphanous1 by Prohibitin2 promotes muscle differentiation. Sci Rep 2019; 9:8302. [PMID: 31165762 PMCID: PMC6549159 DOI: 10.1038/s41598-019-44749-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/23/2019] [Indexed: 02/06/2023] Open
Abstract
Muscle differentiation is controlled by adhesion and growth factor-dependent signalling through common effectors that regulate muscle-specific transcriptional programs. Here we report that mDiaphanous1, an effector of adhesion-dependent RhoA-signalling, negatively regulates myogenesis at the level of Myogenin expression. In myotubes, over-expression of mDia1ΔN3, a RhoA-independent mutant, suppresses Myogenin promoter activity and expression. We investigated mDia1-interacting proteins that may counteract mDia1 to permit Myogenin expression and timely differentiation. Using yeast two-hybrid and mass-spectrometric analysis, we report that mDia1 has a stage-specific interactome, including Prohibitin2, MyoD, Akt2, and β-Catenin, along with a number of proteosomal and mitochondrial components. Of these interacting partners, Prohibitin2 colocalises with mDia1 in cytoplasmic punctae in myotubes. We mapped the interacting domains of mDia1 and Phb2, and used interacting (mDia1ΔN3/Phb2 FL or mDia1ΔN3/Phb2-Carboxy) and non-interacting pairs (mDia1H + P/Phb2 FL or mDia1ΔN3/Phb2-Amino) to dissect the functional consequences of this partnership on Myogenin promoter activity. Co-expression of full-length as well as mDia1-interacting domains of Prohibitin2 reverse the anti-myogenic effects of mDia1ΔN3, while non-interacting regions do not. Our results suggest that Prohibitin2 sequesters mDia1, dampens its anti-myogenic activity and fine-tunes RhoA-mDia1 signalling to promote differentiation. Overall, we report that mDia1 is multi-functional signalling effector whose anti-myogenic activity is modulated by a differentiation-dependent interactome. The data have been deposited to the ProteomeXchange with identifier PXD012257.
Collapse
Affiliation(s)
- Amena Saleh
- Institute for Stem Cell Science & Regenerative Medicine, Bangalore, Karnataka, 560065, India
- Council of Scientific & Industrial Research -Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India
- Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Gunasekaran Subramaniam
- Council of Scientific & Industrial Research -Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Swasti Raychaudhuri
- Council of Scientific & Industrial Research -Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India
| | - Jyotsna Dhawan
- Institute for Stem Cell Science & Regenerative Medicine, Bangalore, Karnataka, 560065, India.
- Council of Scientific & Industrial Research -Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India.
| |
Collapse
|
24
|
Castiglioni I, Caccia R, Garcia-Manteiga JM, Ferri G, Caretti G, Molineris I, Nishioka K, Gabellini D. The Trithorax protein Ash1L promotes myoblast fusion by activating Cdon expression. Nat Commun 2018; 9:5026. [PMID: 30487570 PMCID: PMC6262021 DOI: 10.1038/s41467-018-07313-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 10/24/2018] [Indexed: 12/17/2022] Open
Abstract
Myoblast fusion (MF) is required for muscle growth and repair, and its alteration contributes to muscle diseases. The mechanisms governing this process are incompletely understood, and no epigenetic regulator has been previously described. Ash1L is an epigenetic activator belonging to the Trithorax group of proteins and is involved in FSHD muscular dystrophy, autism and cancer. Its physiological role in skeletal muscle is unknown. Here we report that Ash1L expression is positively correlated with MF and reduced in Duchenne muscular dystrophy. In vivo, ex vivo and in vitro experiments support a selective and evolutionary conserved requirement for Ash1L in MF. RNA- and ChIP-sequencing indicate that Ash1L is required to counteract Polycomb repressive activity to allow activation of selected myogenesis genes, in particular the key MF gene Cdon. Our results promote Ash1L as an important epigenetic regulator of MF and suggest that its activity could be targeted to improve cell therapy for muscle diseases. Myoblast fusion in skeletal muscle is a complex process but how this is regulated is unclear. Here, the authors identify Ash1L, a histone methyltransferase, as modulating myoblast fusion via activation of the myogenesis gene Cdon, and observe decreased Ash1L expression in Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Ilaria Castiglioni
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy
| | - Roberta Caccia
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy
| | - Jose Manuel Garcia-Manteiga
- Center for Translational Genomics and BioInformatics, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy
| | - Giulia Ferri
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy
| | - Giuseppina Caretti
- Department of Biosciences, University of Milan, via Celoria 26, Milano, 20133, Italy
| | - Ivan Molineris
- Center for Translational Genomics and BioInformatics, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy
| | - Kenichi Nishioka
- Department of Biomolecular Sciences, Division of Molecular Genetics and Epigenetics, Faculty of Medicine, Saga University, Saga, Japan.,Laboratory for Developmental Genetics, RIKEN IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan
| | - Davide Gabellini
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy.
| |
Collapse
|
25
|
Hamoud N, Tran V, Aimi T, Kakegawa W, Lahaie S, Thibault MP, Pelletier A, Wong GW, Kim IS, Kania A, Yuzaki M, Bouvier M, Côté JF. Spatiotemporal regulation of the GPCR activity of BAI3 by C1qL4 and Stabilin-2 controls myoblast fusion. Nat Commun 2018; 9:4470. [PMID: 30367035 PMCID: PMC6203814 DOI: 10.1038/s41467-018-06897-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 10/05/2018] [Indexed: 11/09/2022] Open
Abstract
Myoblast fusion is tightly regulated during development and regeneration of muscle fibers. BAI3 is a receptor that orchestrates myoblast fusion via Elmo/Dock1 signaling, but the mechanisms regulating its activity remain elusive. Here we report that mice lacking BAI3 display small muscle fibers and inefficient muscle regeneration after cardiotoxin-induced injury. We describe two proteins that repress or activate BAI3 in muscle progenitors. We find that the secreted C1q-like1-4 proteins repress fusion by specifically interacting with BAI3. Using a proteomic approach, we identify Stabilin-2 as a protein that interacts with BAI3 and stimulates its fusion promoting activity. We demonstrate that Stabilin-2 activates the GPCR activity of BAI3. The resulting activated heterotrimeric G-proteins contribute to the initial recruitment of Elmo proteins to the membrane, which are then stabilized on BAI3 through a direct interaction. Collectively, our results demonstrate that the activity of BAI3 is spatiotemporally regulated by C1qL4 and Stabilin-2 during myoblast fusion.
Collapse
Affiliation(s)
- Noumeira Hamoud
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, H2W 1R7, Canada.,Département de Médecine (Programmes de Biologie Moléculaire), Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Viviane Tran
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, H2W 1R7, Canada.,Département de Biochimie, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Takahiro Aimi
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JT), Tokyo, 102-0075, Japan
| | - Wataru Kakegawa
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JT), Tokyo, 102-0075, Japan
| | - Sylvie Lahaie
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, H2W 1R7, Canada.,Integrated Program in Neuroscience, McGill University, Montréal, QC, H3A 2B4, Canada
| | - Marie-Pier Thibault
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, H2W 1R7, Canada
| | - Ariane Pelletier
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, H2W 1R7, Canada
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.,Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - In-San Kim
- Biomedical Research Institute, Korea Institute Science and Technology, Seoul, 136-791, Republic of Korea.,KU-KIST school, Korea University, Seoul, 136-701, Republic of Korea
| | - Artur Kania
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, H2W 1R7, Canada.,Integrated Program in Neuroscience, McGill University, Montréal, QC, H3A 2B4, Canada.,Department of Anatomy and Cell Biology, McGill University, Montréal, QC, H3A 1A3, Canada
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JT), Tokyo, 102-0075, Japan
| | - Michel Bouvier
- Département de Biochimie, Université de Montréal, Montréal, QC, H3T 1J4, Canada.,Institut de Recherches en Immunologie et Cancérologie (IRIC), Université de Montréal, Montréal, QC, Canada, H3C 3J7
| | - Jean-François Côté
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, H2W 1R7, Canada. .,Département de Médecine (Programmes de Biologie Moléculaire), Université de Montréal, Montréal, QC, H3T 1J4, Canada. .,Département de Biochimie, Université de Montréal, Montréal, QC, H3T 1J4, Canada. .,Department of Anatomy and Cell Biology, McGill University, Montréal, QC, H3A 1A3, Canada.
| |
Collapse
|
26
|
Planchon D, Rios Morris E, Genest M, Comunale F, Vacher S, Bièche I, Denisov EV, Tashireva LA, Perelmuter VM, Linder S, Chavrier P, Bodin S, Gauthier-Rouvière C. MT1-MMP targeting to endolysosomes is mediated by upregulation of flotillins. J Cell Sci 2018; 131:jcs.218925. [PMID: 30111578 DOI: 10.1242/jcs.218925] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/21/2018] [Indexed: 12/31/2022] Open
Abstract
Tumor cell invasion and metastasis formation are the major cause of death in cancer patients. These processes rely on extracellular matrix (ECM) degradation mediated by organelles termed invadopodia, to which the transmembrane matrix metalloproteinase MT1-MMP (also known as MMP14) is delivered from its reservoir, the RAB7-containing endolysosomes. How MT1-MMP is targeted to endolysosomes remains to be elucidated. Flotillin-1 and -2 are upregulated in many invasive cancers. Here, we show that flotillin upregulation triggers a general mechanism, common to carcinoma and sarcoma, which promotes RAB5-dependent MT1-MMP endocytosis and its delivery to RAB7-positive endolysosomal reservoirs. Conversely, flotillin knockdown in invasive cancer cells greatly reduces MT1-MMP accumulation in endolysosomes, its subsequent exocytosis at invadopodia, ECM degradation and cell invasion. Our results demonstrate that flotillin upregulation is necessary and sufficient to promote epithelial and mesenchymal cancer cell invasion and ECM degradation by controlling MT1-MMP endocytosis and delivery to the endolysosomal recycling compartment.
Collapse
Affiliation(s)
- Damien Planchon
- CRBM, Univ Montpellier, CNRS, France, 1919 Route de Mende, 34293 Montpellier, France
| | - Eduardo Rios Morris
- CRBM, Univ Montpellier, CNRS, France, 1919 Route de Mende, 34293 Montpellier, France
| | - Mallory Genest
- CRBM, Univ Montpellier, CNRS, France, 1919 Route de Mende, 34293 Montpellier, France
| | - Franck Comunale
- CRBM, Univ Montpellier, CNRS, France, 1919 Route de Mende, 34293 Montpellier, France
| | - Sophie Vacher
- Department of Genetics, Institut Curie, 75005 Paris, France
| | - Ivan Bièche
- Department of Genetics, Institut Curie, 75005 Paris, France
| | - Evgeny V Denisov
- Cancer Research Institute, Tomsk National Research Medical Center, Tomsk 634050, Russia.,Tomsk State University, Tomsk 634050, Russia
| | - Lubov A Tashireva
- Cancer Research Institute, Tomsk National Research Medical Center, Tomsk 634050, Russia
| | - Vladimir M Perelmuter
- Cancer Research Institute, Tomsk National Research Medical Center, Tomsk 634050, Russia
| | - Stefan Linder
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, University Medical Center Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Philippe Chavrier
- Cell Dynamics and Compartmentalization Unit, Institut Curie, 75005 Paris, France
| | - Stéphane Bodin
- CRBM, Univ Montpellier, CNRS, France, 1919 Route de Mende, 34293 Montpellier, France
| | | |
Collapse
|
27
|
COMP-Angiopoietin-1 accelerates muscle regeneration through N-cadherin activation. Sci Rep 2018; 8:12323. [PMID: 30120297 PMCID: PMC6098079 DOI: 10.1038/s41598-018-30513-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/27/2018] [Indexed: 11/17/2022] Open
Abstract
Angiopoietin-1 modulates vascular stability via Tie2 on endothelial cells. In our previous study, we also showed it acts as an inhibitor of cardiomyocyte death. However, it remains poorly understood how Ang1 regulates myogenesis during muscle regeneration. Here we found that COMP-Ang1 (cAng1) enhances muscle regeneration through N-cadherin activation. Muscle fiber regeneration after limb muscle damage by ischemic injury was enhanced with cAng1 treatment. Mechanistically cAng1 directly bound to N-cadherin on the myoblast surface in a Ca2+ dependent manner. The interaction enhanced N-cadherin activation via N-cadherin/p120-catenin complex formation, which in turn activated p38MAPK (but not AKT or ERK) and myogenin expression (but not myoD) as well as increasing myogenin+ cells in/ex vivo. After transplantation of GFP-expressing myoblasts (GFP-MB), we showed an increased generation of GFP+ myotubes with adenovirus cAng1 (Adv-cAng1) injection. Adv-cAng1, however, could not stimulate myotube formation in N-cadherin-depleted GFP-MB. Taken together, this study uncovers the mechanism of how cAng1 promotes myoblast differentiation and muscle regeneration through the N-cadherin/p120-catenin/p38MAPK/myogenin axis.
Collapse
|
28
|
Somers SM, Spector AA, DiGirolamo DJ, Grayson WL. Biophysical Stimulation for Engineering Functional Skeletal Muscle. TISSUE ENGINEERING PART B-REVIEWS 2018; 23:362-372. [PMID: 28401807 DOI: 10.1089/ten.teb.2016.0444] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tissue engineering is a promising therapeutic strategy to regenerate skeletal muscle. However, ex vivo cultivation methods typically result in a low differentiation efficiency of stem cells as well as grafts that resemble the native tissues morphologically, but lack contractile function. The application of biomimetic tensile strain provides a potent stimulus for enhancing myogenic differentiation and engineering functional skeletal muscle grafts. We reviewed integrin-dependent mechanisms that potentially link mechanotransduction pathways to the upregulation of myogenic genes. Yet, gaps in our understanding make it challenging to use these pathways to theoretically determine optimal ex vivo strain regimens. A multitude of strain protocols have been applied to in vitro cultures for the cultivation of myogenic progenitors (adipose- and bone marrow-derived stem cells and satellite cells) and transformed murine myoblasts, C2C12s. Strain regimens are characterized by orientation, amplitude, and time-dependent factors (effective frequency, duration, and the rest period between successive strain cycles). Analysis of published data has identified possible minimum/maximum values for these parameters and suggests that uniaxial strains may be more potent than biaxial strains, possibly because they more closely mimic physiologic strain profiles. The application of these biophysical stimuli for engineering 3D skeletal muscle grafts is nontrivial and typically requires custom-designed bioreactors used in combination with biomaterial scaffolds. Consideration of the physical properties of these scaffolds is critical for effective transmission of the applied strains to encapsulated cells. Taken together, these studies demonstrate that biomimetic tensile strain generally results in improved myogenic outcomes in myogenic progenitors and differentiated myoblasts. However, for 3D systems, the optimization of the strain regimen may require the entire system including cells, biomaterials, and bioreactor, to be considered in tandem.
Collapse
Affiliation(s)
- Sarah M Somers
- 1 Department of Biomedical Engineering, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Translational Tissue Engineering Center, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Alexander A Spector
- 1 Department of Biomedical Engineering, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Translational Tissue Engineering Center, Johns Hopkins University School of Medicine , Baltimore, Maryland.,3 Institute for Nanobiotechnology (INBT), Johns Hopkins University Whiting School of Engineering , Baltimore, Maryland
| | - Douglas J DiGirolamo
- 4 Department of Orthopedics, Johns Hopkins University School of Medicine , Baltimore Maryland
| | - Warren L Grayson
- 1 Department of Biomedical Engineering, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Translational Tissue Engineering Center, Johns Hopkins University School of Medicine , Baltimore, Maryland.,3 Institute for Nanobiotechnology (INBT), Johns Hopkins University Whiting School of Engineering , Baltimore, Maryland.,5 Department of Material Sciences and Engineering, Johns Hopkins University , Whiting School of Engineering, Baltimore, Maryland
| |
Collapse
|
29
|
Mu X, Tang Y, Takayama K, Chen W, Lu A, Wang B, Weiss K, Huard J. RhoA/ROCK inhibition improves the beneficial effects of glucocorticoid treatment in dystrophic muscle: implications for stem cell depletion. Hum Mol Genet 2018; 26:2813-2824. [PMID: 28549178 DOI: 10.1093/hmg/ddx117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/10/2017] [Indexed: 12/31/2022] Open
Abstract
Glucocorticoid treatment represents a standard palliative treatment for Duchenne muscular dystrophy (DMD) patients, but various adverse effects have limited this treatment. In an effort to understand the mechanism(s) by which glucocorticoids impart their effects on the dystrophic muscle, and potentially reduce the adverse effects, we have studied the effect of prednisolone treatment in dystrophin/utrophin double knockout (dKO) mice, which exhibit a severe dystrophic phenotype due to rapid muscle stem cell depletion. Our results indicate that muscle stem cell depletion in dKO muscle is related to upregulation of mTOR, and that prednisolone treatment reduces the expression of mTOR and other pro-inflammatory mediators, consequently slowing down muscle stem cell depletion. However, prednisolone treatment was unable to improve the myogenesis of stem cells and reduce fibrosis in dKO muscle. We then studied whether glucocorticoid treatment can be improved by co-administration of an inhibitor of RhoA/ROCK signaling, which can be activated by glucocorticoids and was found in our previous work to be over-activated in dystrophic muscle. Our results indicate that the combination of RhoA/ROCK inhibition and glucocorticoid treatment in dystrophic muscle have a synergistic effect in alleviating the dystrophic phenotype. Taken together, our study not only shed light on the mechanism by which glucocorticoid imparts its beneficial effect on dystrophic muscle, but also revealed the synergistic effect of RhoA/ROCK inhibition and glucocorticoid treatment, which could lead to the development of more efficient therapeutic approaches for treating DMD patients.
Collapse
Affiliation(s)
- Xiaodong Mu
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Ying Tang
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Koji Takayama
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Wanqun Chen
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,Department of Biochemistry and Molecular Biology, Jinan University, Guangdong, China
| | - Aiping Lu
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Bing Wang
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Kurt Weiss
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| |
Collapse
|
30
|
Deng S, Azevedo M, Baylies M. Acting on identity: Myoblast fusion and the formation of the syncytial muscle fiber. Semin Cell Dev Biol 2017; 72:45-55. [PMID: 29101004 DOI: 10.1016/j.semcdb.2017.10.033] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/25/2017] [Accepted: 10/30/2017] [Indexed: 12/25/2022]
Abstract
The study of Drosophila muscle development dates back to the middle of the last century. Since that time, Drosophila has proved to be an ideal system for studying muscle development, differentiation, function, and disease. As in humans, Drosophila muscle forms via a series of conserved steps, starting with muscle specification, myoblast fusion, attachment to tendon cells, interactions with motorneurons, and sarcomere and myofibril formation. The genes and mechanisms required for these processes share striking similarities to those found in humans. The highly tractable genetic system and imaging approaches available in Drosophila allow for an efficient interrogation of muscle biology and for application of what we learn to other systems. In this article, we review our current understanding of muscle development in Drosophila, with a focus on myoblast fusion, the process responsible for the generation of syncytial muscle cells. We also compare and contrast those genes required for fusion in Drosophila and vertebrates.
Collapse
Affiliation(s)
- Su Deng
- Program in Developmental Biology, Sloan Kettering Institute, New York, NY 10065, United States
| | - Mafalda Azevedo
- Program in Developmental Biology, Sloan Kettering Institute, New York, NY 10065, United States; Graduate Program in Basic and Applied Biology (GABBA), Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Mary Baylies
- Program in Developmental Biology, Sloan Kettering Institute, New York, NY 10065, United States.
| |
Collapse
|
31
|
Yue B, Wu J, Wang Y, Zhang C, Fang X, Chen H. Expression Profiles Analysis and Functional Characterization of MicroRNA-660 in Skeletal Muscle Differentiation. J Cell Biochem 2017; 118:2387-2394. [PMID: 28106300 DOI: 10.1002/jcb.25901] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/18/2017] [Indexed: 12/17/2022]
Abstract
MicroRNA are a series of small non-coding RNAs that have emerged as critical regulators of skeletal muscle development. Here, we concentrated on the function of miR-660 during bovine skeletal myogenesis from our previous high-throughput sequencing results, then analyzed its expression profiles and characterized related functional roles. Overexpression of miR-660 significantly attenuated myogenic differentiation of C2C12 cells, whereas miR-660 inhibition enhanced C2C12 differentiation. Dual-Luciferase Reporter Assay went for demonstrating that miR-660 directly targeted the 3'-UTR of Rho guanine nucleotide exchange factor 12 (ARHGEF-12). Furthermore, we found an inverse relationship between the expression of miR-660 and ARHGEF12 in both gain- and loss-of-function studies: overexpression of miR-660 declined the mRNA and protein expressions of ARHGEF12 in C2C12 cells differentiation; however, knockdown of miR-660 had completely opposite results. Taken together, these results offered a new perspective for miR-660 in skeletal muscle differentiation. J. Cell. Biochem. 118: 2387-2394, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Binglin Yue
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, P.R. China
| | - Jiyao Wu
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, P.R. China
| | - Yanhuan Wang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, P.R. China
| | - Chunlei Zhang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, P.R. China
| | - Xingtang Fang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, P.R. China
| | - Hong Chen
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, P.R. China
| |
Collapse
|
32
|
Friedl P, Mayor R. Tuning Collective Cell Migration by Cell-Cell Junction Regulation. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a029199. [PMID: 28096261 DOI: 10.1101/cshperspect.a029199] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Collective cell migration critically depends on cell-cell interactions coupled to a dynamic actin cytoskeleton. Important cell-cell adhesion receptor systems implicated in controlling collective movements include cadherins, immunoglobulin superfamily members (L1CAM, NCAM, ALCAM), Ephrin/Eph receptors, Slit/Robo, connexins and integrins, and an adaptive array of intracellular adapter and signaling proteins. Depending on molecular composition and signaling context, cell-cell junctions adapt their shape and stability, and this gradual junction plasticity enables different types of collective cell movements such as epithelial sheet and cluster migration, branching morphogenesis and sprouting, collective network migration, as well as coordinated individual-cell migration and streaming. Thereby, plasticity of cell-cell junction composition and turnover defines the type of collective movements in epithelial, mesenchymal, neuronal, and immune cells, and defines migration coordination, anchorage, and cell dissociation. We here review cell-cell adhesion systems and their functions in different types of collective cell migration as key regulators of collective plasticity.
Collapse
Affiliation(s)
- Peter Friedl
- Department of Cell Biology, Radboud University Medical Centre, Nijmegen 6525GA, The Netherlands.,David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030.,Cancer Genomics Center, 3584 CG Utrecht, The Netherlands
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
33
|
Controlled Heat Stress Promotes Myofibrillogenesis during Myogenesis. PLoS One 2016; 11:e0166294. [PMID: 27824934 PMCID: PMC5100975 DOI: 10.1371/journal.pone.0166294] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 10/26/2016] [Indexed: 11/24/2022] Open
Abstract
Hyperthermia therapy has recently emerged as a clinical modality used to finely tune heat stress inside the human body for various biomedical applications. Nevertheless, little is known regarding the optimal timing or temperature of heat stress that is needed to achieve favorable results following hyperthermia therapy for muscle regeneration purposes. The regeneration of skeletal muscle after injury is a highly complex and coordinated process that involves a multitude of cellular mechanisms. The main objective of this study was to characterize the effects of hyperthermal therapy on the overall behavior of myoblasts during myogenic differentiation. Various cellular processes, including myogenesis, myofibrillogenesis, hypertrophy/atrophy, and mitochondrial biogenesis, were studied using systematic cellular, morphological, and pathway-focused high-throughput gene expression profiling analyses. We found that C2C12 myoblasts exhibited distinctive time and temperature-dependence in biosynthesis and regulatory events during myogenic differentiation. Specifically, we for the first time observed that moderate hyperthermia at 39°C favored the growth of sarcomere in myofibrils at the late stage of myogenesis, showing universal up-regulation of characteristic myofibril proteins. Characteristic myofibrillogenesis genes, including heavy polypeptide 1 myosin, heavy polypeptide 2 myosin, alpha 1 actin, nebulin and titin, were all significantly upregulated (p<0.01) after C2C12 cells differentiated at 39°C over 5 days compared with the control cells cultured at 37°C. Furthermore, moderate hyperthermia enhanced myogenic differentiation, with nucleus densities per myotube showing 2.2-fold, 1.9-fold and 1.6-fold increases when C2C12 cells underwent myogenic differentiation at 39°C over 24 hours, 48 hours and 72 hours, respectively, as compared to the myotubes that were not exposed to heat stress. Yet, atrophy genes were sensitive even to moderate hyperthermia, indicating that strictly controlled heat stress is required to minimize the development of atrophy in myotubes. In addition, mitochondrial biogenesis was enhanced following thermal induction of myoblasts, suggesting a subsequent shift toward anabolic demand requirements for energy production. This study offers a new perspective to understand and utilize the time and temperature-sensitive effects of hyperthermal therapy on muscle regeneration.
Collapse
|
34
|
Díaz-Núñez M, Díez-Torre A, De Wever O, Andrade R, Arluzea J, Silió M, Aréchaga J. Histone deacetylase inhibitors induce invasion of human melanoma cells in vitro via differential regulation of N-cadherin expression and RhoA activity. BMC Cancer 2016; 16:667. [PMID: 27549189 PMCID: PMC4994393 DOI: 10.1186/s12885-016-2693-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/09/2016] [Indexed: 01/11/2023] Open
Abstract
Background Histone deacetylase inhibitors (HDACi) exert multiple cytotoxic actions on cancer cells. Currently, different synthetic HDACi are in clinical use or clinical trials; nevertheless, since both pro-invasive and anti-invasive activities have been described, there is some controversy about the effect of HDACi on melanoma cells. Methods Matrigel and Collagen invasion assays were performed to evaluate the effect of several HDACi (Butyrate, Trichostatin A, Valproic acid and Vorinostat) on two human melanoma cell line invasion (A375 and HT-144). The expression of N- and E-Cadherin and the activity of the RhoA GTPase were analyzed to elucidate the mechanisms involved in the HDACi activity. Results HDACi showed a pro-invasive effect on melanoma cells in vitro. This effect was accompanied by an up-regulation of N-cadherin expression and an inhibition of RhoA activity. Moreover, the down-regulation of N-cadherin through blocking antibodies or siRNA abrogated the pro-invasive effect of the HDACi and, additionally, the inhibition of the Rho/ROCK pathway led to an increase of melanoma cell invasion similar to that observed with the HDACi treatments. Conclusion These results suggest a role of N-cadherin and RhoA in HDACi induced invasion and call into question the suitability of some HDACi as antitumor agents for melanoma patients.
Collapse
Affiliation(s)
- María Díaz-Núñez
- Laboratory of Stem Cells, Development & Cancer, Department of Cell Biology & Histology, Faculty of Medicine & Nursing, University of the Basque Country (UPV/EHU), Leioa, Biscay, Spain
| | - Alejandro Díez-Torre
- Analytical & High Resolution Biomedical Microscopy Core Facility, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Radiotherapy and Experimental Cancer Research, Ghent University Hospital, Ghent, Belgium
| | - Ricardo Andrade
- Analytical & High Resolution Biomedical Microscopy Core Facility, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Jon Arluzea
- Laboratory of Stem Cells, Development & Cancer, Department of Cell Biology & Histology, Faculty of Medicine & Nursing, University of the Basque Country (UPV/EHU), Leioa, Biscay, Spain.,Analytical & High Resolution Biomedical Microscopy Core Facility, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Margarita Silió
- Laboratory of Stem Cells, Development & Cancer, Department of Cell Biology & Histology, Faculty of Medicine & Nursing, University of the Basque Country (UPV/EHU), Leioa, Biscay, Spain
| | - Juan Aréchaga
- Laboratory of Stem Cells, Development & Cancer, Department of Cell Biology & Histology, Faculty of Medicine & Nursing, University of the Basque Country (UPV/EHU), Leioa, Biscay, Spain. .,Analytical & High Resolution Biomedical Microscopy Core Facility, University of the Basque Country (UPV/EHU), Leioa, Spain. .,Department of Cell Biology & Histology, Faculty of Medicine & Dentistry, University of the Basque Country, E-48940, Leioa, Spain.
| |
Collapse
|
35
|
Gribova V, Liu CY, Nishiguchi A, Matsusaki M, Boudou T, Picart C, Akashi M. Construction and myogenic differentiation of 3D myoblast tissues fabricated by fibronectin-gelatin nanofilm coating. Biochem Biophys Res Commun 2016; 474:515-521. [PMID: 27125461 DOI: 10.1016/j.bbrc.2016.04.130] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 04/24/2016] [Indexed: 11/16/2022]
Abstract
In this study, we used a recently developed approach of coating the cells with fibronectin-gelatin nanofilms to build 3D skeletal muscle tissue models. We constructed the microtissues from C2C12 myoblasts and subsequently differentiated them to form muscle-like tissue. The thickness of the constructs could be successfully controlled by altering the number of seeded cells. We were able to build up to ∼76 μm thick 3D constructs that formed multinucleated myotubes. We also found that Rho-kinase inhibitor Y27632 improved myotube formation in thick constructs. Our approach makes it possible to rapidly form 3D muscle tissues and is promising for the in vitro construction of physiologically relevant human skeletal muscle tissue models.
Collapse
Affiliation(s)
- Varvara Gribova
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Chun-Yen Liu
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Akihiro Nishiguchi
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Thomas Boudou
- Grenoble Institute of Technology, CNRS UMR 5628, Laboratory of Materials and Physical Engineering, 3 Parvis L. Néel, 38016 Grenoble, France
| | - Catherine Picart
- Grenoble Institute of Technology, CNRS UMR 5628, Laboratory of Materials and Physical Engineering, 3 Parvis L. Néel, 38016 Grenoble, France.
| | - Mitsuru Akashi
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
36
|
Abstract
Mammalian life begins with a cell-cell fusion event, i.e. the fusion of the spermatozoid with the oocyte and needs further cell-cell fusion processes for the development, growth, and maintenance of tissues and organs over the whole life span. Furthermore, cellular fusion plays a role in infection, cancer, and stem cell-dependent regeneration as well as including an expanded meaning of partial cellular fusion, nanotube formation, and microparticle-cell fusion. The cellular fusion process is highly regulated by proteins which carry the information to organize and regulate membranes allowing the merge of two separate lipid bilayers into one. The regulation of this genetically and epigenetically controlled process is achieved by different kinds of signals leading to communication of fusing cells. The local cellular and extracellular environment additionally initiates specific cell signaling necessary for the induction of the cell-cell fusion process. Common motifs exist in distinct cell-cell fusion processes and their regulation. However, there is specific regulation of different cell-cell fusion processes, e.g. myoblast, placental, osteoclast, and stem cell fusion. Hence, specialized fusion events vary between cell types and species. Molecular mechanisms remain largely unknown, especially limited knowledge is present for cancer and stem cell fusion mechanisms and regulation. More research is necessary for the understanding of cellular fusion processes which can lead to development of new therapeutic strategies grounding on cellular fusion regulation.
Collapse
Affiliation(s)
- Lena Willkomm
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933, Cologne, Germany
| | | |
Collapse
|
37
|
Grefte S, Adjobo-Hermans M, Versteeg E, Koopman W, Daamen W. Impaired primary mouse myotube formation on crosslinked type I collagen films is enhanced by laminin and entactin. Acta Biomater 2016; 30:265-276. [PMID: 26555376 DOI: 10.1016/j.actbio.2015.11.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 10/16/2015] [Accepted: 11/06/2015] [Indexed: 02/05/2023]
Abstract
In skeletal muscle, the stem cell niche is important for controlling the quiescent, proliferation and differentiation states of satellite cells, which are key for skeletal muscle regeneration after wounding. It has been shown that type I collagen, often used as 3D-scaffolds for regenerative medicine purposes, impairs myoblast differentiation. This is most likely due to the absence of specific extracellular matrix proteins providing attachment sites for myoblasts and/or myotubes. In this study we investigated the differentiation capacity of primary murine myoblasts on type I collagen films either untreated or modified with elastin, laminin, type IV collagen, laminin/entactin complex, combinations thereof, and Matrigel as a positive control. Additionally, increased reactive oxygen species (ROS) and ROCK signaling might also be involved. To measure ROS levels with live-cell microscopy, fibronectin-coated glass coverslips were additionally coated with type I collagen and Matrigel onto which myoblasts were differentiated. On type I collagen-coated coverslips, myotube formation was impaired while ROS levels were increased. However, anti-oxidant treatment did not enhance myotube formation. ROCK inhibition, which generally improve cellular attachment to uncoated surfaces or type I collagen, enhanced myoblast attachment to type I collagen-coated coverslips and -films, but slightly enhanced myotube formation. Only modification of type I collagen films by Matrigel and a combination of laminin/entactin significantly improved myotube formation. Our results indicate that type I collagen scaffolds can be modified by satellite cell niche factors of which specifically laminin and entactin enhanced myotube formation. This offers a promising approach for regenerative medicine purposes to heal skeletal muscle wounds. STATEMENT OF SIGNIFICANCE In this manuscript we show for the first time that impaired myotube formation on type I collagen scaffolds can be completely restored by modification with laminin and entactin, two extracellular proteins from the satellite cell niche. This offers a promising approach for regenerative medicine approaches to heal skeletal muscle wounds.
Collapse
|
38
|
Ozawa M. E-cadherin cytoplasmic domain inhibits cell surface localization of endogenous cadherins and fusion of C2C12 myoblasts. Biol Open 2015; 4:1427-35. [PMID: 26453620 PMCID: PMC4728358 DOI: 10.1242/bio.013938] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Myoblast fusion is a highly regulated process that is essential for skeletal muscle formation during muscle development and regeneration in mammals. Much remains to be elucidated about the molecular mechanism of myoblast fusion although cadherins, which are Ca(2+)-dependent cell-cell adhesion molecules, are thought to play a critical role in this process. Mouse myoblasts lacking either N-cadherin or M-cadherin can still fuse to form myotubes, indicating that they have no specific function in this process and may be functionally replaced by either M-cadherin or N-cadherin, respectively. In this study, we show that expressing the E-cadherin cytoplasmic domain ectopically in C2C12 myoblasts inhibits cell surface localization of endogenous M-cadherin and N-cadherin, as well as cell-cell fusion. This domain, however, does not inhibit myoblast differentiation according to microarray-based gene expression analysis. In contrast, expressing a dominant-negative β-catenin mutant ectopically, which suppresses Wnt/β-catenin signaling, did not inhibit cell-cell fusion. Therefore, the E-cadherin cytoplasmic domain inhibits cell-cell fusion by inhibiting cell surface localization of endogenous cadherins and not by inhibiting Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Masayuki Ozawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| |
Collapse
|
39
|
Deshpande RS, Grayson WL, Spector AA. A Modeling Insight into Adipose-Derived Stem Cell Myogenesis. PLoS One 2015; 10:e0137918. [PMID: 26378788 PMCID: PMC4574760 DOI: 10.1371/journal.pone.0137918] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 08/23/2015] [Indexed: 12/02/2022] Open
Abstract
Adipose-derived stem cells (ASCs) are clinically important in regenerative medicine as they are relatively easy to obtain, are characterized by low morbidity, and can differentiate into myogenic progenitor cells. Although studies have elucidated the principal markers, PAX7, Desmin, MyoD, and MHC, the underlying mechanisms are not completely understood. This motivates the application of computational methods to facilitate greater understanding of such processes. In the following, we present a multi-stage kinetic model comprising a system of ordinary differential equations (ODEs). We sought to model ASC differentiation using data from a static culture, where no strain is applied, and a dynamic culture, where 10% strain is applied. The coefficients of the equations have been modulated by those experimental data points. To correctly represent the trajectories, various switches and a feedback factor based on total cell number have been introduced to better represent the biology of ASC differentiation. Furthermore, the model has then been applied to predict ASC fate for strains different from those used in the experimental conditions and for times longer than the duration of the experiment. Analysis of the results reveals unique characteristics of ASC myogenesis under dynamic conditions of the applied strain.
Collapse
Affiliation(s)
- Rajiv S. Deshpande
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Warren L. Grayson
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Alexander A. Spector
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
40
|
Garcia de la Serrana D, Devlin RH, Johnston IA. RNAseq analysis of fast skeletal muscle in restriction-fed transgenic coho salmon (Oncorhynchus kisutch): an experimental model uncoupling the growth hormone and nutritional signals regulating growth. BMC Genomics 2015; 16:564. [PMID: 26228074 PMCID: PMC4521378 DOI: 10.1186/s12864-015-1782-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/15/2015] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Coho salmon (Oncorhynchus kisutch) transgenic for growth hormone (Gh) express Gh in multiple tissues which results in increased appetite and continuous high growth with satiation feeding. Restricting Gh-transgenics to the same lower ration (TR) as wild-type fish (WT) results in similar growth, but with the recruitment of fewer, larger diameter, muscle skeletal fibres to reach a given body size. In order to better understand the genetic mechanisms behind these different patterns of muscle growth and to investigate how the decoupling of Gh and nutritional signals affects gene regulation we used RNA-seq to compare the fast skeletal muscle transcriptome in TR and WT coho salmon. RESULTS Illumina sequencing of individually barcoded libraries from 6 WT and 6 TR coho salmon yielded 704,550,985 paired end reads which were used to construct 323,115 contigs containing 19,093 unique genes of which >10,000 contained >90 % of the coding sequence. Transcripts coding for 31 genes required for myoblast fusion were identified with 22 significantly downregulated in TR relative to WT fish, including 10 (vaspa, cdh15, graf1, crk, crkl, dock1, trio, plekho1a, cdc42a and dock5) associated with signaling through the cell surface protein cadherin. Nineteen out of 44 (43 %) translation initiation factors and 14 of 47 (30 %) protein chaperones were upregulated in TR relative to WT fish. CONCLUSIONS TR coho salmon showed increased growth hormone transcripts and gene expression associated with protein synthesis and folding than WT fish even though net rates of protein accretion were similar. The uncoupling of Gh and amino acid signals likely results in additional costs of transcription associated with protein turnover in TR fish. The predicted reduction in the ionic costs of homeostasis in TR fish associated with increased fibre size were shown to involve multiple pathways regulating myotube fusion, particularly cadherin signaling.
Collapse
Affiliation(s)
| | - Robert H Devlin
- Department of Fisheries and Oceans, Centre for Aquaculture and Environmental Research, 4160 Marine Drive, West Vancouver, BC, V7V 1N6, Canada.
| | - Ian A Johnston
- Scottish Oceans Institute, School of Biology, University of St Andrews, KY16 8LB, St Andrews, Scotland, UK.
| |
Collapse
|
41
|
Grefte S, Wagenaars JAL, Jansen R, Willems PHGM, Koopman WJH. Rotenone inhibits primary murine myotube formation via Raf-1 and ROCK2. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1606-14. [PMID: 25827955 DOI: 10.1016/j.bbamcr.2015.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 03/04/2015] [Accepted: 03/19/2015] [Indexed: 11/30/2022]
Abstract
Rotenone (ROT) is a widely used inhibitor of complex I (CI), the first complex of the mitochondrial oxidative phosphorylation (OXPHOS) system. However, particularly at high concentrations ROT was also described to display off-target effects. Here we studied how ROT affected in vitro primary murine myotube formation. We demonstrate that myotube formation is specifically inhibited by ROT (10-100nM), but not by piericidin A (PA; 100nM), another CI inhibitor. At 100nM, both ROT and PA fully blocked myoblast oxygen consumption. Knock-down of Rho-associated, coiled-coil containing protein kinase 2 (ROCK2) and, to a lesser extent ROCK1, prevented the ROT-induced inhibition of myotube formation. Moreover, the latter was reversed by inhibiting Raf-1 activity. In contrast, ROT-induced inhibition of myotube formation was not prevented by knock-down of RhoA. Taken together, our results support a model in which ROT reduces primary myotube formation independent of its inhibitory effect on CI-driven mitochondrial ATP production, but via a mechanism primarily involving the Raf-1/ROCK2 pathway.
Collapse
Affiliation(s)
- Sander Grefte
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jori A L Wagenaars
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Renate Jansen
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter H G M Willems
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Werner J H Koopman
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
42
|
Kirby TJ, Chaillou T, McCarthy JJ. The role of microRNAs in skeletal muscle health and disease. Front Biosci (Landmark Ed) 2015; 20:37-77. [PMID: 25553440 DOI: 10.2741/4298] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the last decade non-coding RNAs have emerged as importance regulators of gene expression. In particular, microRNAs are a class of small RNAs of ∼ 22 nucleotides that repress gene expression through a post-transcriptional mechanism. MicroRNAs have been shown to be involved in a broader range of biological processes, both physiological and pathological, including myogenesis, adaptation to exercise and various myopathies. The purpose of this review is to provide a comprehensive summary of what is currently known about the role of microRNAs in skeletal muscle health and disease.
Collapse
Affiliation(s)
- Tyler J Kirby
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA, 2Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Thomas Chaillou
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA, 2Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - John J McCarthy
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA, 2Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
43
|
Sumida GM, Yamada S. Rho GTPases and the downstream effectors actin-related protein 2/3 (Arp2/3) complex and myosin II induce membrane fusion at self-contacts. J Biol Chem 2014; 290:3238-47. [PMID: 25527498 DOI: 10.1074/jbc.m114.612168] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Actin regulation is required for membrane activities that drive cell adhesion and migration. The Rho GTPase family plays critical roles in actin and membrane dynamics; however, the roles of the Rho GTPase family are not limited to cell adhesion and migration. Using micron-sized obstacles to induce the formation of self-contacts in epithelial cells, we previously showed that self-adhesion is distinct from cell-to-cell adhesion in that self-contacts are eliminated by membrane fusion. In the current study, we identified Rho GTPases, RhoA, Rac1, and Cdc42, as potential upstream regulators of membrane fusion. The RhoA downstream effector myosin II is required for fusion as the expression of mutant myosin light chain reduced membrane fusion. Furthermore, an inhibitor of the Arp2/3 complex, a downstream effector of Rac1 and Cdc42, also reduced self-contact-induced membrane fusion. At self-contacts, while the concentration of E-cadherin diminished, the intensity of GFP-tagged Arp3 rapidly fluctuated then decreased and stabilized after membrane fusion. Taken together, these data suggest that the Arp2/3 complex-mediated actin polymerization brings two opposing membranes into close apposition by possibly excluding E-cadherin from contact sites, thus promoting membrane fusion at self-contacts.
Collapse
Affiliation(s)
- Grant M Sumida
- From the Department of Biomedical Engineering, University of California, Davis, California 95616
| | - Soichiro Yamada
- From the Department of Biomedical Engineering, University of California, Davis, California 95616
| |
Collapse
|
44
|
Singh RK, Xia Z, Bland CS, Kalsotra A, Scavuzzo MA, Curk T, Ule J, Li W, Cooper TA. Rbfox2-coordinated alternative splicing of Mef2d and Rock2 controls myoblast fusion during myogenesis. Mol Cell 2014; 55:592-603. [PMID: 25087874 PMCID: PMC4142074 DOI: 10.1016/j.molcel.2014.06.035] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 05/15/2014] [Accepted: 06/27/2014] [Indexed: 12/29/2022]
Abstract
Alternative splicing plays important regulatory roles during periods of physiological change. During development, a large number of genes coordinately express protein isoform transitions regulated by alternative splicing; however, the mechanisms that coordinate splicing and the functional integration of the resultant tissue-specific protein isoforms are typically unknown. Here we show that the conserved Rbfox2 RNA binding protein regulates 30% of the splicing transitions observed during myogenesis and is required for the specific step of myoblast fusion. Integration of Rbfox2-dependent splicing outcomes from RNA-seq with Rbfox2 iCLIP data identified Mef2d and Rock2 as Rbfox2 splicing targets. Restored activities of Mef2d and Rock2 rescued myoblast fusion in Rbfox2-depleted cultures, demonstrating functional cooperation of protein isoforms generated by coordinated alterative splicing. The results demonstrate that coordinated alternative splicing by a single RNA binding protein modulates transcription (Mef2d) and cell signaling (Rock2) programs to drive tissue-specific functions (cell fusion) to promote a developmental transition.
Collapse
Affiliation(s)
- Ravi K. Singh
- Departments of Pathology and Immunology, Baylor College of Medicine, Medicine, Houston, Texas 77030, USA
| | - Zheng Xia
- Departments of Molecular and Cellular Biology, Baylor College of Medicine, Medicine, Houston, Texas 77030, USA
- Departments of Dan L. Duncan Cancer Center, Baylor College of Medicine, Medicine, Houston, Texas 77030, USA
| | - Christopher S. Bland
- The Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Medicine, Houston, Texas 77030, USA
| | - Auinash Kalsotra
- Departments of Pathology and Immunology, Baylor College of Medicine, Medicine, Houston, Texas 77030, USA
| | - Marissa A. Scavuzzo
- Departments of Pathology and Immunology, Baylor College of Medicine, Medicine, Houston, Texas 77030, USA
| | - Tomaz Curk
- University of Ljubljana, Faculty of Computer and Information Science, Trẑaŝka cesta 25, SI-1000 Ljubljana
| | - Jernej Ule
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Wei Li
- Departments of Molecular and Cellular Biology, Baylor College of Medicine, Medicine, Houston, Texas 77030, USA
- Departments of Dan L. Duncan Cancer Center, Baylor College of Medicine, Medicine, Houston, Texas 77030, USA
| | - Thomas A. Cooper
- Departments of Pathology and Immunology, Baylor College of Medicine, Medicine, Houston, Texas 77030, USA
- Departments of Molecular and Cellular Biology, Baylor College of Medicine, Medicine, Houston, Texas 77030, USA
- Departments of Molecular Physiology and Biophysics, Baylor College of Medicine, Medicine, Houston, Texas 77030, USA
| |
Collapse
|
45
|
Vafiadaki E, Arvanitis DA, Papalouka V, Terzis G, Roumeliotis TI, Spengos K, Garbis SD, Manta P, Kranias EG, Sanoudou D. Muscle lim protein isoform negatively regulates striated muscle actin dynamics and differentiation. FEBS J 2014; 281:3261-79. [PMID: 24860983 DOI: 10.1111/febs.12859] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 05/14/2014] [Accepted: 05/22/2014] [Indexed: 11/28/2022]
Abstract
Muscle lim protein (MLP) has emerged as a critical regulator of striated muscle physiology and pathophysiology. Mutations in cysteine and glycine-rich protein 3 (CSRP3), the gene encoding MLP, have been directly associated with human cardiomyopathies, whereas aberrant expression patterns are reported in human cardiac and skeletal muscle diseases. Increasing evidence suggests that MLP has an important role in both myogenic differentiation and myocyte cytoarchitecture, although the full spectrum of its intracellular roles has not been delineated. We report the discovery of an alternative splice variant of MLP, designated as MLP-b, showing distinct expression in neuromuscular disease and direct roles in actin dynamics and muscle differentiation. This novel isoform originates by alternative splicing of exons 3 and 4. At the protein level, it contains the N-terminus first half LIM domain of MLP and a unique sequence of 22 amino acids. Physiologically, it is expressed during early differentiation, whereas its overexpression reduces C2C12 differentiation and myotube formation. This may be mediated through its inhibition of MLP/cofilin-2-mediated F-actin dynamics. In differentiated striated muscles, MLP-b localizes to the sarcomeres and binds directly to Z-disc components, including α-actinin, T-cap and MLP. The findings of the present study unveil a novel player in muscle physiology and pathophysiology that is implicated in myogenesis as a negative regulator of myotube formation, as well as in differentiated striated muscles as a contributor to sarcomeric integrity.
Collapse
Affiliation(s)
- Elizabeth Vafiadaki
- Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, Greece
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
p120 modulates LPS-induced NF-κB activation partially through RhoA in bronchial epithelial cells. BIOMED RESEARCH INTERNATIONAL 2014; 2014:932340. [PMID: 24995336 PMCID: PMC4065672 DOI: 10.1155/2014/932340] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 04/22/2014] [Accepted: 05/08/2014] [Indexed: 12/03/2022]
Abstract
p120-Catenin (p120) is an adherens junction protein recognized to regulate cell-cell adhesion. Emerging evidence indicates that p120 may also play an important role in inflammatory responses, and the regulatory mechanisms are still unknown. In the present study, we showed that p120 was associated with airway inflammation. p120 downregulation induced nuclear factor-κB (NF-κB) activation, accompanied with IκBα degradation, p65 nuclear translocation, and increased expression of interleukin-8 (IL-8) in lipopolysaccharide (LPS)- treated C57BL mice and human bronchial epithelial cells (BECs). Moreover, we first found that p120 directly coprecipitated with RhoA in BECs. After LPS stimulation, although total RhoA and p120-bound RhoA were unchanged, RhoA activity was increased. Y27632, a ROCK inhibitor, could partially inhibit nuclear translocation of p65. Overexpression of p120 inactivated RhoA and NF-κB in BECs, whereas p120 loss significantly increased RhoA activity, p65 nuclear translocation, and IL-8 expression. Taken together, our study supports the regulatory role of p120 in airway inflammation and reveals that p120 may modulate NF-κB signaling partially through RhoA.
Collapse
|
47
|
Alexander MS, Casar JC, Motohashi N, Vieira NM, Eisenberg I, Marshall JL, Gasperini MJ, Lek A, Myers JA, Estrella EA, Kang PB, Shapiro F, Rahimov F, Kawahara G, Widrick JJ, Kunkel LM. MicroRNA-486-dependent modulation of DOCK3/PTEN/AKT signaling pathways improves muscular dystrophy-associated symptoms. J Clin Invest 2014; 124:2651-67. [PMID: 24789910 DOI: 10.1172/jci73579] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by mutations in the gene encoding dystrophin, which results in dysfunctional signaling pathways within muscle. Previously, we identified microRNA-486 (miR-486) as a muscle-enriched microRNA that is markedly reduced in the muscles of dystrophin-deficient mice (Dmdmdx-5Cv mice) and in DMD patient muscles. Here, we determined that muscle-specific transgenic overexpression of miR-486 in muscle of Dmdmdx-5Cv mice results in reduced serum creatine kinase levels, improved sarcolemmal integrity, fewer centralized myonuclei, increased myofiber size, and improved muscle physiology and performance. Additionally, we identified dedicator of cytokinesis 3 (DOCK3) as a miR-486 target in skeletal muscle and determined that DOCK3 expression is induced in dystrophic muscles. DOCK3 overexpression in human myotubes modulated PTEN/AKT signaling, which regulates muscle hypertrophy and growth, and induced apoptosis. Furthermore, several components of the PTEN/AKT pathway were markedly modulated by miR-486 in dystrophin-deficient muscle. Skeletal muscle-specific miR-486 overexpression in Dmdmdx-5Cv animals decreased levels of DOCK3, reduced PTEN expression, and subsequently increased levels of phosphorylated AKT, which resulted in an overall beneficial effect. Together, these studies demonstrate that stable overexpression of miR-486 ameliorates the disease progression of dystrophin-deficient skeletal muscle.
Collapse
MESH Headings
- Animals
- Base Sequence
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Line
- Guanine Nucleotide Exchange Factors/genetics
- Guanine Nucleotide Exchange Factors/metabolism
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Mice, Transgenic
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Animal/genetics
- Muscular Dystrophy, Animal/metabolism
- Muscular Dystrophy, Animal/pathology
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- PTEN Phosphohydrolase/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Sequence Homology, Nucleic Acid
- Signal Transduction
- Up-Regulation
Collapse
|
48
|
Lamon S, Wallace MA, Russell AP. The STARS signaling pathway: a key regulator of skeletal muscle function. Pflugers Arch 2014; 466:1659-71. [DOI: 10.1007/s00424-014-1475-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 02/04/2014] [Accepted: 02/05/2014] [Indexed: 01/08/2023]
|
49
|
Guillaume E, Comunale F, Do Khoa N, Planchon D, Bodin S, Gauthier-Rouvière C. Flotillin microdomains stabilize cadherins at cell-cell junctions. J Cell Sci 2013; 126:5293-304. [PMID: 24046456 DOI: 10.1242/jcs.133975] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cadherins are essential in many fundamental processes and assemble at regions of cell-cell contact in large macromolecular complexes named adherens junctions. We have identified flotillin 1 and 2 as new partners of the cadherin complexes. We show that flotillins are localised at cell-cell junctions (CCJs) in a cadherin-dependent manner. Flotillins and cadherins are constitutively associated at the plasma membrane and their colocalisation at CCJ increases with CCJ maturation. Using three-dimensional structured illumination super-resolution microscopy, we found that cadherin and flotillin complexes are associated with F-actin bundles at CCJs. The knockdown of flotillins dramatically affected N- and E-cadherin recruitment at CCJs in mesenchymal and epithelial cell types and perturbed CCJ integrity and functionality. Moreover, we determined that flotillins are required for cadherin association with GM1-containing plasma membrane microdomains. This allows p120 catenin binding to the cadherin complex and its stabilization at CCJs. Altogether, these data demonstrate that flotillin microdomains are required for cadherin stabilization at CCJs and for the formation of functional CCJs.
Collapse
Affiliation(s)
- Emilie Guillaume
- Equipe Labellisée Ligue Contre le Cancer, Universités Montpellier 2 et 1, CRBM, CNRS, UMR 5237, 1919 Route de Mende, 34293 Montpellier, France
| | | | | | | | | | | |
Collapse
|
50
|
Mukai A, Hashimoto N. Regulation of pre-fusion events: recruitment of M-cadherin to microrafts organized at fusion-competent sites of myogenic cells. BMC Cell Biol 2013; 14:37. [PMID: 23978243 PMCID: PMC3846853 DOI: 10.1186/1471-2121-14-37] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 08/22/2013] [Indexed: 01/16/2023] Open
Abstract
Background Previous research indicates that the membrane ruffles and leading edge of lamellipodia of myogenic cells contain presumptive fusion sites. A micrometer-sized lipid raft (microraft) is organized at the presumptive fusion site of mouse myogenic cells in a cell-contact independent way and serves as a platform tethering adhesion proteins that are relevant to cell fusion. However, the mechanisms underlying recruitment of adhesion proteins to lipid rafts and microraft organization remain unknown. Results Here we show that small G-protein Rac1 was required for microraft organization and subsequent cell fusion. However, Rac1 activity was unnecessary for recruitment of M-cadherin to lipid rafts. We found that p120 catenin (p120) binds to M-cadherin exclusively in lipid rafts of differentiating myogenic cells. The Src kinase inhibitor SU6656 prevented p120 binding to M-cadherin and their recruitment to lipid rafts, then suppressed microraft organization, membrane ruffling, and myogenic cell fusion. Suppression of membrane ruffling in SU6656-treated cells was partially restored by pretreatment with the protein tyrosine phosphatase inhibitor vanadate. The present analyses using an antibody to tyrosine phosphorylated p120 suggest that Src family kinases play a role in binding of p120 to M-cadherin and the recruitment of M-cadherin to lipid rafts through phosphorylation of putative substrates other than p120. Conclusions The present study showed that the procedure establishing fusion-competent sites consists of two sequential events: recruitment of adhesion complexes to lipid rafts and organization of microrafts. The recruitment of M-cadherin to lipid rafts depended on interaction with p120 catenin, whereas the organization of microrafts was controlled by a small G protein, Rac1.
Collapse
Affiliation(s)
- Atsushi Mukai
- Department of Regenerative Medicine, National Institute for Longevity Sciences, National Center for Geriatrics and Gerontology, 35 Gengo, Morioka, Oobu, Aichi 474-8522, Japan.
| | | |
Collapse
|